US5684143A - Oligo-2'-fluoronucleotide N3'->P5' phosphoramidates - Google Patents

Oligo-2'-fluoronucleotide N3'->P5' phosphoramidates Download PDF

Info

Publication number
US5684143A
US5684143A US08/603,566 US60356696A US5684143A US 5684143 A US5684143 A US 5684143A US 60356696 A US60356696 A US 60356696A US 5684143 A US5684143 A US 5684143A
Authority
US
United States
Prior art keywords
sub
oligo
phosphoramidates
polynucleotide
sup
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
US08/603,566
Inventor
Sergei Gryaznov
Ronald G. Schultz
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Geron Corp
Original Assignee
Lynx Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to US08/603,566 priority Critical patent/US5684143A/en
Application filed by Lynx Therapeutics Inc filed Critical Lynx Therapeutics Inc
Priority to CZ982629A priority patent/CZ262998A3/en
Priority to AT96919449T priority patent/ATE212997T1/en
Priority to PCT/US1996/010418 priority patent/WO1997031009A1/en
Priority to US08/663,918 priority patent/US5824793A/en
Priority to CA 2245666 priority patent/CA2245666C/en
Priority to PL96328639A priority patent/PL183661B1/en
Priority to EP96919449A priority patent/EP0882059B1/en
Priority to DE69619141T priority patent/DE69619141T2/en
Priority to AU61789/96A priority patent/AU703509C/en
Priority to US08/771,789 priority patent/US5859233A/en
Application granted granted Critical
Publication of US5684143A publication Critical patent/US5684143A/en
Assigned to LYNX THERAPEUTICS, INC. reassignment LYNX THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SCHULTZ, RONALD G., GRYAZNOV, SERGEI M.
Assigned to GERON CORPORATION reassignment GERON CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LYNX THERAPEUTICS, INC.
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • C07H19/10Pyrimidine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals
    • C07H19/20Purine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids

Definitions

  • the invention relates generally to nucleic acid polymer chemistry, and more particularly, to oligo-2'-fluoronucleotide N3' ⁇ P5' phosphoramidates, methods of synthesizing such compounds, and uses thereof.
  • Nucleic acid polymer chemistry has played a crucial role in many developing technologies, particularly in the pharmaceutical, diagnostic, and analytical fields, and more particularly in the subfields of antisense and anti-gene therapeutics, combinatorial chemistry, branched DNA signal amplification, and array-based DNA diagnostics and analysis e.g. Uhlmann and Peyman, Chemical Reviews, 90:543-584 (1990); Milligan et at, J. Med. Chem. 36:1923-1937 (1993); Mesmaeker et at, Current Opinion in Structural Biology, 5: 343-355 (1995); Thuong et at, Angew. Chem. Int. Ed.
  • PNAs peptide nucleic acids
  • phorphorothioates display good nuclease resistance and solubility, but are typically synthesized as P-chiral mixtures and display several sequence-non-specific biological effects, e.g.
  • methylphosphonates display good nuclease resistance and cellular uptake, but are also typically synthesized as P-chiral mixtures and have reduce duplex stability, e.g. Mesmaeker et al (cited above); and so on.
  • an important objective of our invention is to provide oligonucleotide N3' ⁇ P5' phosphoramidate compounds that have increased acid stability and at the same time retain favorable binding and nuclease resistance properties.
  • Another objective of our invention is to provide a class of compounds that are capable of forming Watson-Crick duplexes of greater stability and of comparable specificity to that of natural DNAs and RNAs.
  • Yet another objective of our invention is to provide a class of compounds that are capable of forming triplex structures with Watson-Crick duplexes of DNA and other nucleic acid polymers.
  • a further objective of our invention is to provide compounds that can be advantageously used in techniques requiting the formation of stable and specific duplex and/or triplex structures, including antisense and/or anti-gene pharmaceuticals, branched DNA components, DNA and/or RNA capture agents, and components of DNA-based diagnostic assays.
  • oligo-2'-fluoronucleoside N3' ⁇ P5' phosphoramidates Preferably, oligo-2'-fluoronucleoside N3' ⁇ P5' phosphoramidates of the invention have the following formula: ##STR1## wherein B is a purine or pyrimidine, R 1 is hydroxyl or a 5'-hydroxyl protecting group, R 2 is hydroxyl or amino, R 3 is hydrogen or fluorine, R 4 is hydrogen or a phosphate protecting group, and n is an integer greater than 1.
  • oligo-2'-fluoronucleoside N3' ⁇ P5' phosphoramidates of the invention are synthesized on solid phase supports using monomers of the invention.
  • synthesis takes place by nucleotide-by-nucleotide addition to a growing oligo-2'-fluoronucleotide N3' ⁇ P5' phosphoramidate chain such that the order and identity of the nucleotide monomer added at each step is controlled. That is, the sequence of B's and the length of the oligomer synthesized is predetermined.
  • FIG. 1 illustrates a scheme for synthesizing a first type uracil monomer for assembling oligo-2'-fluoronucleoside N3' ⁇ P5' phosphoramidates of the invention.
  • FIG. 2 illustrates a scheme for synthesizing a second type uracil monomer for assembling oligo-2'-fluoronucleoside N3' ⁇ P5' phosphoramidates of the invention.
  • FIG. 3 illustrates a scheme for synthesizing cytosine monomers for assembling oligo-2'-fluoronucleoside N3' ⁇ P5' phosphoramidates of the invention.
  • FIG. 4 illustrates a scheme for synthesizing oligo-2'-fluoronucleoside N3' ⁇ P5' phosphoramidates of the invention.
  • FIG. 5 is a 19 F NMR spectrum of 5'-DMT-O-2'-fluoro-3'-amino-2',3'-dideoxyuridine synthesized in accordance with the invention.
  • FIG. 6 is an ion exchange HPLC chromatogram of the crude reaction mixture from synthesis of an oligo-2'-fluoronucleoside N3' ⁇ P5' phosphoramidate of Example 5.
  • oligonucleotide is represented by a sequence of letters, such as "ATGUCCTG,” it will be understood that the nucleotides are in 5' ⁇ 3' order from left to right and that "A” denotes deoxyadenosine, “C” denotes deoxycytidine, “G” denotes deoxyguanosine, “T” denotes thymidine, and “U” denotes deoxyuridine, unless otherwise noted.
  • N3' ⁇ P5' phosphoramidate refers to an internucleosidic linkage of the form:
  • R 4 is hydrogen or a phosphate protecting group. More particularly, R 4 may by alkyl, alkenyl, aryl, aralkyl, or cycloalkyl containing up to 10 carbon atoms.
  • R 4 is alkyl having from 1 to 6 carbon atoms; electron- withdrawing ⁇ -substituted ethyl, particularly ⁇ -trihalomethyl-, ⁇ -cyano-, ⁇ -sulfo-, ⁇ -nitro- substituted ethyl, or the like; electron-withdrawing substituted phenyl, particularly halo-, sulfo-, cyano-, or nitro-, substituted phenyl; or electron-withdrawing substituted phenylethyl. More preferably, R 4 is methyl, ⁇ -cyanoethyl, or 4- nitrophenylethyl.
  • R 4 is hydrogen, methyl, or ⁇ -cyanoethyl.
  • Electron-withdrawing substituents are typically halo, cyano, nitro, sulfo, or mono-, di-, or trihalomethyl, and the like.
  • Halogen atom substituents are usually fluoro, chloro, bromo, or iodo; and preferably, they are fluoro or chloro.
  • Electron-withdrawing denotes the tendancy of a substituent to attract valence electrons of the molecule of which it is apart, i.e. it is electronegative, e.g. March, Advanced Organic Chemistry, pgs. 16-18 (John Wiley, New York, 1985).
  • nucleotide phosphoramidates are sometimes indicated herein by a subscripted “np” or “pn” for N3' ⁇ P5' phosphoramidates and P3' ⁇ N5' phosphoramidates, respectively.
  • "U np U” is a dinucleotide in which a 3'-aminouridine and a deoxyuridine are linked by an N3' ⁇ P5' phosphoramidate linkage.
  • 2'-fluoro substituents are indicated by a superscripted "f".
  • U f np U is a dinucleotide in which the 3'-most 2'-fluorouridine is linked to a uridine by an N3' ⁇ P5' phosphoramidate linkage.
  • a single leading subscripted “p” indicates to a 5' monophosphate, and a single trailing subscripted "n” indicates a 3'-amino.
  • nucleoside includes the natural nucleosides, including 2'-deoxy and 2'-hydroxyl forms, e.g. as described in Kornberg and Baker, DNA Replication, 2nd Ed. (Freeman, San Francisco, 1992).
  • "Analogs" in reference to nucleosides includes synthetic nucleosides having modified base moieties and/or modified sugar moieties, e.g. described generally by Scheit, Nucleotide Analogs (John Wiley, New York, 1980). Such analogs include synthetic nucleosides designed to enhance binding properties, e.g. stability, specificity, or the like, such as disclosed by Uhlmann and Peyman (cited above).
  • pyrimidine means the pyrimidine occurring in natural nucleosides, including cytosine, thymine, and uracil, and common analogs thereof, such as those containing oxy, methyl, methoxy, hydroxyl, amino, thio, and like, substituents.
  • the term as used herein further includes pyrimidines with common protection groups attached, such as 4N-benzoylcytosine. Further common pyrimidine protection groups are disclosed by Beaucage and Iyer, Tetrahedron, 48:2223-23 11 (1992).
  • purine means the purines occurring in natural nucleosides, including adenine, guanine, and hypoxanthine, and common analogs thereof, such as those containing oxy, methyl, methoxy, hydroxyl, amino, thio, and like, substituents.
  • the term as used herein further includes purines with common protection groups attached, such as 2N-benzoylguanine, 2N-isobutrylguanine, 6N-benzoyladenine, and the like. Further common purine protection groups are disclosed by Beaucage and Iyer (cited above).
  • the invention includes polymers of 2'-fluoro-3'-aminonucleosides, that is, oligo-2'-fluoronucleotide N3' ⁇ P5' phosphoramidates, monomers and methods for solid phase assembly of such polymers, and uses of the polymers.
  • the invention also includes compositions comprising Watson-Crick duplexes of RNA or DNA and oligo-2'-fluoronucleotide N3' ⁇ P5' phosphoramidates and compositions comprising triplexes of DNA duplexes and oligo-2'-fluoronucleotide N3' ⁇ P5' phosphoramidates, and methods and kits for using oligo-2'-fluoronucleotide N3' ⁇ P5' phosphoramidates as DNA capture agents based on either Watson-Crick basepairing with single stranded target nucleic acids or Hoogsteen or reverse Hoogsteen triplet formation with double stranded target nucleic acids.
  • oligo-2'-fluoronucleotide N3' ⁇ P5' phosphoramidates of the invention are between 2 and 30 nucleotides in length. More preferably, they are between 8 and 25 nucleotides in length; and most preferably, they are between 8 and 20 nucleotides in length.
  • Oligomers of the invention may be synthesized by several different approaches.
  • a first approach coupling is achieved by carbon tetrachloride-driven oxidative phosphorylation of a nucleoside 3 '-amine by a 5'-H-phosphonate of the terminal nucleotide of a growing chain anchored to a solid phase support, as described, for example, by Gryaznov et al, J. Am. Chem. Soc. 116:3143-3144 (1994); and Chen et al, Nucleic Acids Research, 23: 2661-2668 (1995); which references are incorporated by reference.
  • Monomers for this synthetic approach have the following form: ##STR2## where B is defined as above and R 1 is hydrogen or a hydroxyl protecting group, such as triphenylmethyl (i.e., trityl), p-anisyldiphenylmethyl (i.e., monomethoxytrityl or MMT), di-p-anisylphenylmethyl (i.e., dimethoxytrityl or DMT), pivaloyl, acetyl, 4-methoxytetrahydropyran-4-yl, tetrahydropyranyl, phenoxyacetyl, isobutyloxycarbonyl, pixyl, benzyl, trialkylsilyl having from 3 to 9 carbon atoms, 9-fluorenylmethyl carbamate (Fmoc), or the like.
  • R 1 is DMT.
  • FIG. 1 A general scheme for preparing the above monomers is shown in FIG. 1. Briefly, a ribonucleoside is transformed into a 5'-hydroxyl-protected-2',3'-anhydroxylnucleoside, after which the 2',3'-epoxy ring is opened by treatment with sodium azide, or like reagent, to form a 5'-hydroxyl-protected-3'-azido-3'-deoxyarabinonucleoside.
  • the 5'-hydroxyl-protected-3'-azido-3'-deoxyarabinonucleoside is fluorinated at the 2' position by treatment with diethylaminosulfur trifluoride (DAST), or like reagent, after which the azido group is reduced to give the above monomer.
  • DAST diethylaminosulfur trifluoride
  • the monomers are then employed for chain assembly essentially as described by Chen et al and Gryaznov et al (cited above).
  • oligomers of the invention are synthesized via an exchange reaction between a phosphoramidite group on a monomer and a 3'-amino of an anchored chain, as illustrated by Scheme 4 in FIG. 4.
  • Monomers for this synthetic approach have the following form: ##STR3## where B and R 1 are defined as above, R 5 is an amino protecting group, and R' and R" taken together with the nitrogen form an alkyl- or arylamino leaving group. More particularly, R' and R" taken separately each are alkyl, aralkyl, cycloalkyl, and cycloalkylalkyl containing up to 10 carbon atoms in total.
  • R' and R" taken separately are alkyl having from 1 to 5 carbon atoms. Most preferably, R' and R" taken separately are isopropyl. R' and R" taken together form an alkylene chain containing up to 5 carbon atoms in the principal chain and a total of up to 10 carbon atoms with both terminal valence bonds of said chain being attached to the nitrogen atom to which R' and R" are attached; or R' and R" when taken together with the nitrogen atom to which they are attached form a saturated nitrogen heterocycle which may contain one or more additional heteroatoms from the group consisting of nitrogen, oxygen, and sulfur.
  • R' and R" taken together and with the nitrogen to which they are attached are pyrrolidino, morpholino, or piperidino. Most preferably, R' and R" taken together and with the nitrogen to which they are attached are morpholino.
  • R 5 is trityl, and more preferably, R 5 is 4-methoxytrityl.
  • Phosphoramidite monomers of the invention are prepared as follows, as illustrated in FIGS. 2 and 3.
  • a ribonucleoside is transformed into a 5'-hydroxyl-protected-2',3'-anhydroxylnucleoside, after which the 2',3'-epoxy ring is opened by treatment with sodium azide, or like reagent, to form a 5'-hydroxyl-protected-3'-azido-3'-deoxyarabinonucleoside.
  • the 5'-hydroxyl-protected-3'-azido-3'-deoxyarabinonucleoside is fluorinated at the 2' position by treatment with diethylaminosulfur trifluoride (DAST), or like reagent, after which the azido group is reduced to give a 3'-amino.
  • DAST diethylaminosulfur trifluoride
  • the nucleoside is phosphitylated at the 5' oxygen to give the crude phorphoramidite monomer.
  • the phosphoramidite monomers are used in the following synthetic cycle:
  • This cycle can be repeated, resulting in oligo-2'-fluoronucleotide N3' ⁇ P5' phosphoramidates after cleavage from the solid support and deprotection with ammonia.
  • solid phase supports may be used with the invention, including microparticles made of controlled pore glass (CPG), highly cross-linked polystyrene, acrylic copolymers, cellulose, nylon, dextran, latex, polyacrolein, and the like, disclosed in the following exemplary references: Meth. Enzymol., Section A, pages 11-147, vol. 44 (Academic Press, New York, 1976); U.S. Pat. Nos. 4,678,814; 4,413,070; and 4,046;720; and Pon, Chapter 19, in Agrawal, editor, Methods in Molecular Biology, Vol. 20, (Humana Press, Totowa, N.J., 1993).
  • CPG controlled pore glass
  • Supports further include commercially available nucleoside-derivatized CPG and polystyrene beads (e.g. available from Applied Biosystems, Foster City, Calif.); polystyrene grafted with polyethylene glycol (e.g., TentaGelTM, Rapp Polymere, Tubingen Germany); and the like. Selection of the support characteristics, such as material, porosity, size, shape, and the like, and the type of linking moiety employed depends on a variety of factors, such as protection groups employed, length of final product, quantity of final product, and the like. Exemplary linking moieties are disclosed in Pon et at, Biotechniques, 6:768-775 (1988); Webb, U.S. Pat. No.
  • thermal stability of oligo-2'-fluoronucleotide N3' ⁇ P5' phosphoramidates are determined by way of melting, or strand dissociation, curves.
  • the temperature of fifty percent strand dissociation is taken as the melting temperature, T m , which, in turn, provides a convenient measure of stability.
  • T m measurements are typically carried out in a saline solution at neutral pH with target and oligomer concentrations at between about 1.0-3.0 ⁇ M. Typical conditions are as follows: 150 mM NaCl and 10 mM MgCl 2 in a 10 mM sodium phosphate buffer (pH 7.0) or in a 10 mM Tris-HCl buffer (pH 7.0); or like conditions.
  • Data for melting curves are accumulated by heating a sample, e.g. a duplex or triplex, of the oligo-2'-fluoronucleotide N3' ⁇ P5' phosphoramidate and its complementary single stranded or double stranded oligonucleotide from 10-15° C. to about 85°-90° C.
  • absorbance of 260 nm light is monitored at 1° C. intervals, e.g. using a Cary (Australia) model 1E or a Hewlett-Packard (Palo Alto, Calif.) model HP 8459 UVNIS spectrophotometer and model HP 89100A temperature controller, or like instruments.
  • third strand association via Hoogsteen type of binding is most stable along homopyrimidine-homopufine tracks in a double stranded target.
  • base triplets form in T-A*T or C-G*C motifs (where "-" indicates Watson-Crick pairing and "*" indicates Hoogsteen type of binding); however, other motifs are also possible.
  • Hoogsteen base pairing permits parallel and antiparallel orientations between the third strand (the Hoogsteen strand) and the purine-rich strand of the duplex to which the third strand binds, depending on conditions and the composition of the strands.
  • nucleoside type e.g. whether ribose or deoxyribose nucleosides are employed
  • base modifications e.g. methylated cytosine, and the like
  • Roberts et al Proc. Natl. Acad. Sci., 88:9397-9401 (1991); Roberts et at, Science, 258:1463-1466 (1992); Distefano et al, Proc. Natl. Acad.
  • Compounds of the invention can be employed as diagnostic probes to detect the presence of one or more target polynueleotides in a wide range of samples, including environmental samples, e.g. from public water supplies, samples from foodstuffs, and from other biological samples, such as blood, saliva, semen, amniotic fluid, tissue homogenates of plants or animals, or of human patients, and the like.
  • environmental samples e.g. from public water supplies, samples from foodstuffs, and from other biological samples, such as blood, saliva, semen, amniotic fluid, tissue homogenates of plants or animals, or of human patients, and the like.
  • nucleic acid probes in human diagnostics, forensics, and genetic analysis has been extensively reviewed.
  • Oligo-2'-fluoronucleoside N3' ⁇ P5' phosphoramidates of the invention may be used in essentially any of the known solution or solid phase hybridization formats, such as those in which the analyte is bound directly to a solid phase, or sandwich hybridizations in which the analyte is bound to an oligonucleotide that is, in turn, bound to a solid phase.
  • Oligo-2'-fluoronucleoside N3' ⁇ P5' phosphoramidates are particularly useful in conjunction with branched polymer signal amplification schemes, such as those disclosed by Urdea et al, U.S. Pat. No. 5,124,246; Wang et al, U.S. Pat. No.
  • the oligo-2'-fluoronucleoside N3' ⁇ P5' phosphoramidate preferably serves as a highly stable and specific "capture" probe by binding to a target polynucleotide analyte of interest.
  • Kits incorporating oligo-2'-fluoronucleotide N3' ⁇ P5' phosphoramidates can take a variety of forms depending on the particular embodiment, the type of assay format employed, and the labeling scheme employed. Generally, kits of the invention comprise an oligo-2'-fluoronucleoside N3' ⁇ P5' phosphoramidate specific for a given target polynucleotide, a hybridization buffer, and a signal generation moiety.
  • Kits of the invention may further comprise wash buffers for removing unbound label and/or oligo-2'-fluoronucleoside N3' ⁇ P5' phosphoramidate, solid phase supports such as derivatized magnetic beads, or the like; and prehybridization buffers containing blocking agents, e.g. Denhardt's solution, sonicated salmon sperm DNA, detergents such as 1% SDS, or the like, for minimizing nonspecific binding of the oligo-2'-fluoronucleoside N3' ⁇ P5' phosphoramidate or other nucleosidic binding components, such as amplifier strands.
  • An exemplary hybridization buffer comprises the following reagents: 100-150 mM NaCl, 10 mM MgCl 2 , and 10 mM Tris-HCl (pH 7.0).
  • Compounds of the invention may be used as DNA capture agents by forming highly stable triplex between a double stranded target DNA and an oligo-2'-fluoronucleotide N3' ⁇ P5' phosphoramidate of a predetermined sequence.
  • This application is particularly useful in manipulating multiple double stranded DNAs such as DNA sequencing plasmids of PCR amplicons that contain complementary sequences for binding via triplex formation with the oligo-2'-fluoronucleotide N3' ⁇ P5' phosphoramidates, such as described ordinary oligonucleotides by Cantor et at, U.S. Pat. No. 5,482,836, which patent is incorporated by reference.
  • compounds of the invention are biotinylated or covalently attached to a like binding moiety, such as a hapten.
  • a like binding moiety such as a hapten.
  • the target DNAs are extracted from the sample by contacting the biotin, or like binding moiety, with its receptor which preferably is attached to a solid support, such as a magnetic bead.
  • the captured DNAs may be release under conditions that destroy the stability of the triplex structure, e.g. mild alkaline buffer, pH 8-10.
  • Signal generation moieties are molecular structures that directly or indirectly generate a signal, e.g. fluorescent, colorimetric, radioactive, or the like, that can be detected by conventional means.
  • Direct signal generation means that the moiety producing a signal is covalently linked to the oligo-2'-fluoronucleoside N3' ⁇ P5' phosphoramidate, e.g. as with the covalent attachment of a fluorescent dye, enzyme, or the like.
  • Indirect signal generation means that a particular moiety, e.g. an oligonucleotide tail conjugated to a oligo-2'-fluoronucleoside N3' ⁇ P5' phosphoramidate, is one component of a multi-component system that produces a signal.
  • the signal generation moieties comprises an oligonucleotide tail of about 12 to about 50 nucleotides in length.
  • a signal is generated indirectly by providing a second oligonucleotide which is complementary to the tail and which has a fluorescent dye covalently attached. Attaching fluorescent dyes to oligonucleotides is well known in the art, e.g. U.S. Pat. Nos. 4,997,828; 5,151,507; 4,855,225; 5,188,934; Eckstein, editor (cited above); and the like.
  • compositions of the invention can be employed as antisense or anti-gene compounds for treating disease.
  • the components included in such pharmaceutical compositions of the invention depend on several factors, including the nature of the disease or condition being treated, the location of disease lesions, the mode of drug delivery and/or administration contemplated, the latter of which can include in vivo administration by way of regional or systemic perfusion, topical application, intranasal administration, administration by implanted or transdermal sustained release systems, and the like, as well as ex vivo administration for use in bone marrow purging.
  • a preferred method of administration of oligonucleotides comprises either regional or systemic perfusion.
  • the afferent and efferent vessels supplying the extremity containing a lesion are isolated and connected to a low-flow perfusion pump in continuity with an oxygenator and heat exchanger.
  • the iliac vessels may be used for perfusion of the lower extremities.
  • the axillary vessels are cannulated high in the axilla for upper extremity lesions.
  • a pharmaceutical composition containing an oligo-2'-fluoronucleotide N3' ⁇ P5' phosphoramidate is added to the perfusion circuit, and the perfusion is continued for an appropriate time period, e.g. an hour.
  • Perfusion rates of from 100 to 150 ml/minute may be employed for lower extremity lesions, while half that rate should be employed for upper extremity lesions.
  • Systemic heparinization may be used throughout the perfusion, and reversed after the perfusion is complete. This isolation perfusion technique permits administration of higher dosed of chemotherapeutic agent than would otherwise be tolerated upon infusion into the arterial or venous systemic circulation.
  • a pharmaceutical composition of the invention facilitates the delivery of an effective amount of the active drug to a desired site in a manner consistent with patient safety and comfort.
  • An effective amount of an oligo-2'-fluoronucleotide N3' ⁇ P5' phosphoramidate depends on several factors, including the disease or condition being treated, the method of administration, the scheduling of the administration, the condition of the patient, and the like.
  • a parentially administered dose will be in the range of about 1 g/kg/day to about 100 mg/kg/day of patient body weight.
  • a key factor in selecting an appropriate dose for a given condition or disease is the therapeutic result, as measure by standard criteria well known to the medical practitioner, e.g. for oncological applications see: Cancer: Principles and Practice of Oncology, 3rd Edition, edited by V. T. DeVita et al (Lippincott Company, Philadelphia, 1989).
  • compositions of the invention include a pharmaceutical carrier that may contain a variety of components that provide a variety of functions, including regulation of drug concentration, regulation of solubility, chemical stabilization, regulation of viscosity, absorption enhancement, regulation of pH, and the like.
  • a pharmaceutical carrier that may contain a variety of components that provide a variety of functions, including regulation of drug concentration, regulation of solubility, chemical stabilization, regulation of viscosity, absorption enhancement, regulation of pH, and the like.
  • the pharmaceutical composition preferably includes a buffer such as a phosphate buffer, or other organic acid salt, preferably at a pH of between about 7 and 8.
  • a nonionic surfactant such as Tween 80 in an amount of 0.04-0.05% (w/v), to increase solubility.
  • antioxidants such as ascorbic acid
  • hydrophilic polymers such as, monosaccharides, disaccharides, and other carbohydrates including cellulose or its derivatives, dextrins, chelating agents, such as EDTA, and like components well known to those in the pharmaceutical sciences, e.g. Remington's Pharmaceutical Science, latest edition (Mack Publishing Company, Easton, Pa.).
  • Compounds of the invention include the pharmaceutically acceptable salts thereof, including those of alkaline earths, e.g. sodium or magnesium, ammonium or NX 4 + , wherein X is C 1-4 alkyl.
  • Other pharmaceutically acceptable salts include organic carboxylic acids such as acetic, lactic, tartaric, malic, isethionic, lactobionic, and succinic acids; organic sulfonic acids such as methanesulfonic, ethanesulfonic, and benzenesulfonic; and inorganic acids such as hydrochloric, sulfuric, phosphoric, and sulfamic acids.
  • Pharmaceutically acceptable salts of a compound having a hydroxyl group include the anion of such compound in combination with a suitable cation such as Na + , NH 4 + , or the like.
  • Sustained release systems suitable for use with the pharmaceutical compositons of the invention include semi-permeable polymer matrices in the form of films, microcapsules, or the like, comprising polylactides, copolymers of L-glutamic acid and gamma-ethyl-L-glutamate, poly(2-hydroxyethyl methacrylate), and like materials.
  • Sustained release systems also include liposomally entrapped oligonucleotides, e.g. as described in Liposome Technology, Vol. II, Incorporation of Drugs, Proteins, and Genetic Material (CRC Press).
  • Phosphodiester oligodeoxyribonucleotides and oligoribonucleotides were prepared on an ABI 380B DNA synthesizer using standard protocol via the phosphoramidite method.
  • 18 Oligonucleotide N3' ⁇ P5' phosphoramidates, containing 2'-deoxy- and one or two 2'-fluoronucleosides were synthesized using the oxidative phosphorylation method on a ABI 394 synthesizer as previously described.
  • 4c Uniformly modified oligo-2'-fluoronucleotide N3' ⁇ P5' phosphoramidates were prepared by amidite transfer reaction on an ABI 380B synthesizer using the following protocol:
  • oligonucleotides were analyzed and purified by IE HPLC. Oligonucleotides were desalted on Pharmacia NAP-5 or NAP-10 gel filtration columns immediately after purification and stored frozen or lyophilized at -18° C.
  • Dionex DX300 or DX500 systems were used for IE analysis and purification of oligonucleotides.
  • a Pharmacia MonoQ 10/10 column was used for analysis and purification of crude oligomers, eluted with a 2% per minute gradient of 1.5M NaCl in 10 mM NaOH.
  • a Hewlett Packard Hypersil ODS, 5 g column on a Waters HPLC system was used for RP HPLC, with a 1% per minute gradient of acetonitrile in 0.1M triethylammonium acetate, pH 7.0.
  • NMR spectra were recorded on a Bruker DRX-400 spectrometer. Chemical shifts are reported relative to TMS, CCl 3 F, and H 3 PO 4 , for 1 H, 19 F, and 31 P spectra, respectively.
  • TLC Thin layer chromatography
  • Compound 1, 5'-DMT-2'-fluoro-3'-aminouridine was prepared according to Scheme 1 shown in FIG. 1. This monomer was used for incorporation of 2'-fluoro-3'-aminouridine into oligonucleotide phosphoramidates by the oxidative phosphorylation method. 4 First, uridine 3 was transformed into the 5'-DMT-2',3'-anhydrolyxouridine 4 by successive, one pot reaction with DMT-chloride, mesyl chloride, and sodium hydroxide.
  • the 2',3'-epoxy ring was then opened by treatment with sodium azide, 10 producing 5'-DMT-3'-azido-3'-deoxyarabinouridine 5 as the main product, and isomeric 5'-DMT-2'-azido-2'-deoxyxylouridine as a by-product, in approximately a 2:1 ratio.
  • Compound 5 was isolated by silica gel chromatography and then fluorinated with diethylaminosulfur trifluoride (DAST) to give 6. Finally, the azido group of 6 was reduced with hydrogen over palladium catalyst, giving 5'-DMT-2'-fluoro-3'-aminouridine 1.
  • the structure of nucleoside 1 was confirmed by 1 H and 19 F NMR analysis and by mass spectrometry (FIG. 5). More particularly, the steps were carded out as follows:
  • 5'-0-DMT-2',3'-anhydrolyxouridine (compound 4, FIG. 1) was prepared as follows: To 10.0 g (41 mmol) of dry 3 in 250 mL anhydrous pyridine was added 14.6 g (43 mmol) of dimethoxytrityl chloride. The mixture was stirred overnight, then cooled on ice and 8.0 mL (103 mmol) methanesulfonyl chloride was added. After stirring at room temperature for 4 h, the reaction was quenched with 1 mL H 2 O and the solvent removed in vacuo.
  • 5'-O-DMT-3'-azido-3'-deoxyarabinouridine (compound 5, FIG. 1) was prepared as follows: To 13.8 g (26 mmol) of 4 in 500 mL acetone was added 200 mL H 2 O and 12.0 g (185 mmol) NAN 3 . The mixture was refluxed overnight, then concentrated/n vacuo to remove the acetone. The resultant slurry was extracted with 600 mL CH 2 Cl 2 , which was in turn was washed with water (3 ⁇ 250 mL). Concentration of the CH 2 Cl 2 layer and flash chromatography of the crude product provided 6.0 g (40%) of a pale yellow solid.
  • 5'-O-DMT-2'-fluoro-3'-azido-2',3'-dideoxyuridine (compound 6, FIG. 1) was prepared as follows: To 6.0 g (10.5 mmol) of 5 in 120 mL anhydrous DMF was added 2.4 mL (18.2 mmol) of diethylaminosulfur trifluoride. The mixture was stirred for 16 h, then poured into 300 mL of cold saturated aqueous NaHCO 3 . The product was extracted with 500 mL ethyl acetate, which in turn was washed with water (2 ⁇ 500 mL). Concentration of the organic layer and flash chromatography of the crude product provided 2.9 g (48%) of an off-white solid.
  • 5'-O-DMT-2'-fluoro-3'-amino-2',3'-dideoxyuridine (compound 6, FIG. 1) was prepared as follows: To 2.9 g (5.1 mmol) of 6 in 75 mL of 95% ethanol was added 0.5 g of 10% palladium on carbon. The mixture was hydrogenated at 40 psi overnight and then the catalyst removed by filtration. The solvent was removed in vacuo, and the resultant solid purified by flash chromatography to give 1.2 g (43%) of product as a white powder. Mass-spectrometry, FAB + , M+H + , calculated: 548.2197, observed: 548.2206.
  • Oligomers of the invention were also synthesized from phosphoramidite monomer, as illustrated by compound 2u of Scheme 2.
  • Uridine 3 was mesylated and then selectively benzoylated with accompanying formation of the 2,2'-anhydrocycle by treatment with sodium benzoate according to literature procedure. 11 These reactions resulted in compound 7 with 69-77% overall yields.
  • 12 2,3'-anhydroarabinonueleoside 7 was transformed into 2',3'-anhydrolyxoufidine 8 in two steps.
  • 3'-O-Methanesulfonyl-5'-O-benzoyl-2,2'-anhydroarabinouridine (compound 7, FIG. 2) was prepared in two steps from 3 (FIG. 2) according to the procedure of Codington et al. 11 in 69-77% overall yields.
  • 5'-O-benzoyl-2',3'-anhydrolyxouridine (compound 8, FIG. 2) was prepared in two steps from 7 according to the procedure of Codington et al. 12 in 63-77% overall yields.
  • 3'-azido-5'-O-benzoyl-3'-deoxyarabinouridine (compound 9, FIG. 2) was prepared from 8 and anhydrous NH 4 N 3 , 20 according to the procedure of Reichman et al., 13 but without successful recrystallization. Mass yields were 98% or greater, but NMR suggested 25-35% of the regioisomer, 2'-azido-5'-O-benzoyl-2'-deoxyxylouridine, 9i, which coeluted with the desired product by silica gel TLC.
  • 2'-fluoro-3'-amino-5'-O-benzoyl-2',3'-dideoxyuridine (compound 11, FIG. 2) was prepared as follows: To 3.5 g (9.3 mmol) crude 10 (20% 10i) in 200 mL 95% ethanol was added 600 mg of 10% palladium on carbon. The suspension was hydrogenated at 40 psi overnight and then the catalyst removed by filtration. The solvent was removed in vacuo, giving 2.93 g (90%) of a light yellow solid consisting of two compounds which were resolvable by TLC. Flash chromatography provided 1.96 g (60% yield) of the desired product as a pure white solid.
  • 2'-fluoro-3'-(4-methoxytrityl)amino-2',3'-dideoxyuridine (compound 12, FIG. 2) was prepared as follows: To 1.0 g (2.9 mmol) of 11 in 50 mL anhydrous pyridine was added 1.0 g (3.2 mmol) 4-methoxytrityl chloride. The mixture was stirred overnight, 5 mL saturated aqueous NaHCO 3 was added, and the mixture concentrated in vacuo to an oil. The oil was dissolved in 125 mL ethyl acetate, which was washed with water (3 ⁇ 100 mL) and reconcentrated in vacuo to 2.05 g of foam.
  • N 4 -benzoyl-2'-fluoro-3'-(4-methoxytrityl)amino-2',3'-dideoxyuridine 5'-(2-cyanoethyl N,N-diisopropyl)phosphoramidite (compound 2u, FIG. 2) was prepared as follows: To 700 mg (1.35 mmol) of 12 in 20 mL anhydrous CH 2 Cl 2 was added 200 mg (1.17 mmol) of diisopropylammonium tetrazolide and 0.5 mL (1.57 mmol) of 2-cyanoethyl N,N,N',N'-tetraisopropylphosphorodiamidite.
  • N 4 ,5'-O-dibenzoyl-2'-fluoro-3'-amino-2',3'-dideoxycytidine (compound 13, FIG. 3) was prepared as follows: To 6.9 g (18.4 mmol) of crude 10 (containing 35% 10i) in 50 mL anhydrous CH 3 CN was added an ice-cold solution of 11.7 g (169 mmol) 1,2,4-triazole and 3.35 mL (36.1 mmol) POCl 3 in 90 mL anhydrous CH 3 CN. The mixture was cooled in an ice bath and anhydrous triethylamine (23 mL, 165 mmol) was added, then the reaction allowed to warm to room temperature with stirring.
  • N 4 ,5 '-O-dibenzoyl-2'-fluoro-3'-(4-methoxytrityl) amino-2',3'-dideoxycytidine (compound 14, FIG. 3) was prepared as follows: To 0.9 g (2.0 mmol) of 13 in 25 mL anhydrous pyridine was added 0.86 g (2.8 mmol) 4-methoxytrityl chloride, and the mixture stirred overnight. The reaction was quenched with 0.5 mL H 2 O and concentrated in vacuo. CH 2 Cl 2 (50 mL) was added and washed with 50 mL saturated aqueous NaHCO3 and with water (2 ⁇ 50 mL).
  • N 4 -benzoyl-2'-fluoro-3'-(4-methoxytrityl) amino-2',3'-dideoxycytidine (compound 15, FIG. 3) was prepared as follows: To 1.3 g (1.75 mmol) of 14 in 20 mL of 65/30/5 pyridine/methanol/water, cooled in an ice bath, was added 10 mL of cold 2M NaOH in 65/30/5 pyrridine/methanol/water. The mixture was stirred cold for 20 min, then neutralized with pyridinium-H + form Bio-Rad AG® 50W-X8 cation exchange resin. After 5 min, the resin was removed by filtration and washed with methanol.
  • N 4 -benzoyl-2'-fluoro-3'-(4-methoxytrityl)amino-2',3'-dideoxycytidine 5'-(2-cyanoethyl N,N-diisopropyl)phosphoramidite (compound 2c, FIG. 3) was prepared as follows: To 970 mg (1.56 mmol) of 15 in 25 mL anhydrous CH 2 Cl 2 was added 200 mg (1.17 mmol) of diisopropylammonium tetrazolide and 1.0 mL (3.15 mmol) of 2-cyanoethyl N,N,N',N'-tetraisopropylphosphorodiamidite.
  • the oil was dissolved in 20 mL CH 2 Cl 2 , washed with 20 mL of saturated aqueous NaHCO 3 and with water (2 ⁇ 20mL), and then reconcentrated in vacuo to a foam.
  • To the foam was added 1 mL 0.4M diisopropylethylamine in DMSO/N-methylpyrrolidine, 1/1, and 0.7 mL 0.2M 1-hydroxybenzotriazole, 0.2M 2-(1H-benzotriazol-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate in DMSO/N-methylpyrrolidine, 1/1.
  • Oligo-2'-fluoronucleotide N3' ⁇ P5' phosphoramidates were synthesized on solid phase supports using either 5'-DMT-2'-fluoro-3'-aminonucleoside monomers (Scheme 1) or phosphoramidite monomers (of Schemes 2 and 3).
  • Dimer dU f np T was prepared using monomer of Scheme 1 via carbon tetrachloride-driven oxidative phosphorylation of dU f n by a 5'-H-phosphonate of the anchored dT.
  • Product was analyzed and isolated by RP HPLC in 70% yield and the structure was confirmed by mass spectrometry and by acid catalyzed hydrolysis, which gave 2'-fluoro-3'-amino-2'-deoxyuridine and 5'-thymidylic acid.
  • the same synthetic strategy was also used to introduce one or two 2'-fluoro-3'-aminonucleosides into longer oligonucleotide phosphoramidate chains.
  • T m values were increased by 16°-25° C. (compare expt. 2 and 5, Table 1).
  • the presented data show that further increases in the proportion of N-sugar ring pucker in the N3' ⁇ P5' phosphoramidates as well as additional negative polarization of 3'-amino groups (by 2'-fluorine) substantially stabilized oligonucleotide duplexes. It is noteworthy that T m 's of the duplexes formed by 2'-fluoroamidates were 38°-44° C.
  • oligo-2'-fluoro- Stability of the oligo-2'-fluoro- in comparison with the oligo-2'-deoxynucleotide N3' ⁇ P5' phosphoramidates toward enzymatic hydrolysis was evaluated.
  • Phosphoramidates 19 and 22 (Table 1) were treated with snake venom phosphodiesterase and alkaline phosphatase, and analyzed at successive time points by IE HPLC. Under the conditions used, oligo-2'-deoxyphosphoramidate 19 and oligo-2'-fluorophosphoramidate 22 were hydrolyzed progressively and at similar rates, with calculated half-lives of the full-length strands equal to 4.9 and 5.4 h, respectively. In comparison, decathymidilic acid with natural phosphodiester linkages was completely digested to thymidine within 10 min under the same conditions.
  • the oligo-2'-fluorophosphoramidate 22 was noticeably more stable under these conditions, with respective half-lives of this full-length oligomer of 61 min, 66 h, and 309 h. These results demonstrate a markedly reduced 3'-nitrogen basicity due to electron-withdrawing 2'-fluorine, with consequently greater acid stability of oligo-2'-fluorophosphoramidates relative to the parent oligo-2'-deoxyphosphoramidates.

Abstract

A new class of oligonucleotide N3'→P5' phosphoramidates having 2' fluoro substituents are provided that have superior acid stability. The invention includes oligo-2'-fluoronucleotide N3'→P5' phosphoramidates, methods of synthesis, and duplexes and triplexes formed with DNA and RNA. Compounds of the invention are useful where the formation of stable and specific duplex and/or triplex structures is desired, including antisense and/or anti-gene pharmaceuticals, branched DNA components, DNA and/or RNA capture agents, components of DNA-based diagnostic assays, and the like.

Description

The invention relates generally to nucleic acid polymer chemistry, and more particularly, to oligo-2'-fluoronucleotide N3'→P5' phosphoramidates, methods of synthesizing such compounds, and uses thereof.
BACKGROUND
Nucleic acid polymer chemistry has played a crucial role in many developing technologies, particularly in the pharmaceutical, diagnostic, and analytical fields, and more particularly in the subfields of antisense and anti-gene therapeutics, combinatorial chemistry, branched DNA signal amplification, and array-based DNA diagnostics and analysis e.g. Uhlmann and Peyman, Chemical Reviews, 90:543-584 (1990); Milligan et at, J. Med. Chem. 36:1923-1937 (1993); Mesmaeker et at, Current Opinion in Structural Biology, 5: 343-355 (1995); Thuong et at, Angew. Chem. Int. Ed. Engl., 32:666-690 (1993); Brenner et at, Proc. Natl. Acad. Sci., 89:5381-5383 (1992); Gold et at, Ann. Rev. Biochem., 64: 763-797 (1995); Gallop et at, J. Med. Chem. 37:1233-1258 (1994); Gordon et al, J. Med. Chem. 37: 1385-1401 (1994); Gryaznov, International application PCTFUS94/07557; Urdea et at, U.S. Pat. No. 5,124,246; Southern et at, Genomics, 13:1008-1017 (1992); McGall et at, U.S. Pat. No. 5,412,087; Fodor et at, U.S. Pat. No. 5,424,186; Pirrung et at, U.S. Pat. No. 5,405,783; and the like.
Much of this chemistry has been directed to improving the binding strength, specificity, and nuclease resistance of natural nucleic acid polymers, such as DNA. Unfortunately, improvements in one property, such as nuclease resistance, often involve trade-offs against other properties, such as binding strength. Examples of such trade-offs abound: peptide nucleic acids (PNAs) display good nuclease resistance and binding strength, but have reduced cellular uptake in test cultures, e.g. Hanvey et at, Science, 258: 1481-1485 (1992); phorphorothioates display good nuclease resistance and solubility, but are typically synthesized as P-chiral mixtures and display several sequence-non-specific biological effects, e.g. Stein et at, Science, 261:1004-1012 (1993); methylphosphonates display good nuclease resistance and cellular uptake, but are also typically synthesized as P-chiral mixtures and have reduce duplex stability, e.g. Mesmaeker et al (cited above); and so on.
Recently, a new class of oligonucleotide analog has been developed having so-called N3'→P5' phorphoramidate internucleoside linkages. These compounds display very favorable binding properties, nuclease resistance, and solubility, but like most other phosphoramidates have reduced stability under acidic conditions, Gryaznov and Letsinger, Nucleic Acids Research, 20:3403-3409 (1992); Chen et at, Nucleic Acids Research, 23: 2661-2668 (1995); Gryaznov et at, Proc. Natl. Acad. Sci., 92:5798-5802 (1995); and Gryaznov et al, J. Am. Chem. Soc., 116:3143-3144 (1994).
The utility of this new class of oligonucleotide analog would be significantly increased if modifications could be found that broadened its range of acid stability, without a concomitant loss in any other of its other favorable properties outlined above.
SUMMARY OF THE INVENTION
In view of the above, an important objective of our invention is to provide oligonucleotide N3'→P5' phosphoramidate compounds that have increased acid stability and at the same time retain favorable binding and nuclease resistance properties.
Another objective of our invention is to provide a class of compounds that are capable of forming Watson-Crick duplexes of greater stability and of comparable specificity to that of natural DNAs and RNAs.
Yet another objective of our invention is to provide a class of compounds that are capable of forming triplex structures with Watson-Crick duplexes of DNA and other nucleic acid polymers.
A further objective of our invention is to provide compounds that can be advantageously used in techniques requiting the formation of stable and specific duplex and/or triplex structures, including antisense and/or anti-gene pharmaceuticals, branched DNA components, DNA and/or RNA capture agents, and components of DNA-based diagnostic assays.
These and other objects of our invention are accomplished with oligo-2'-fluoronucleoside N3'→P5' phosphoramidates. Preferably, oligo-2'-fluoronucleoside N3'→P5' phosphoramidates of the invention have the following formula: ##STR1## wherein B is a purine or pyrimidine, R1 is hydroxyl or a 5'-hydroxyl protecting group, R2 is hydroxyl or amino, R3 is hydrogen or fluorine, R4 is hydrogen or a phosphate protecting group, and n is an integer greater than 1.
Preferably, oligo-2'-fluoronucleoside N3'→P5' phosphoramidates of the invention are synthesized on solid phase supports using monomers of the invention. In further preference, synthesis takes place by nucleotide-by-nucleotide addition to a growing oligo-2'-fluoronucleotide N3'→P5' phosphoramidate chain such that the order and identity of the nucleotide monomer added at each step is controlled. That is, the sequence of B's and the length of the oligomer synthesized is predetermined.
BRIEF DESCRIPTION OF THE FIGURES
FIG. 1 illustrates a scheme for synthesizing a first type uracil monomer for assembling oligo-2'-fluoronucleoside N3'→P5' phosphoramidates of the invention.
FIG. 2 illustrates a scheme for synthesizing a second type uracil monomer for assembling oligo-2'-fluoronucleoside N3'→P5' phosphoramidates of the invention.
FIG. 3 illustrates a scheme for synthesizing cytosine monomers for assembling oligo-2'-fluoronucleoside N3'→P5' phosphoramidates of the invention.
FIG. 4 illustrates a scheme for synthesizing oligo-2'-fluoronucleoside N3'→P5' phosphoramidates of the invention.
FIG. 5 is a 19 F NMR spectrum of 5'-DMT-O-2'-fluoro-3'-amino-2',3'-dideoxyuridine synthesized in accordance with the invention.
FIG. 6 is an ion exchange HPLC chromatogram of the crude reaction mixture from synthesis of an oligo-2'-fluoronucleoside N3'→P5' phosphoramidate of Example 5.
DEFINITIONS
Whenever an oligonucleotide is represented by a sequence of letters, such as "ATGUCCTG," it will be understood that the nucleotides are in 5'→3' order from left to right and that "A" denotes deoxyadenosine, "C" denotes deoxycytidine, "G" denotes deoxyguanosine, "T" denotes thymidine, and "U" denotes deoxyuridine, unless otherwise noted.
As used herein, "N3'→P5' phosphoramidate" refers to an internucleosidic linkage of the form:
3'--NH--P(═O)(OR.sup.4)--O-- 5'
wherein the 3' and 5' refer to the carbon atoms of the sugar moieties of consecutive nucleosides which are connected by way of the linkage, and wherein R4 is hydrogen or a phosphate protecting group. More particularly, R4 may by alkyl, alkenyl, aryl, aralkyl, or cycloalkyl containing up to 10 carbon atoms. Preferably, R4 is alkyl having from 1 to 6 carbon atoms; electron- withdrawing β-substituted ethyl, particularly β-trihalomethyl-, β-cyano-, β-sulfo-, β-nitro- substituted ethyl, or the like; electron-withdrawing substituted phenyl, particularly halo-, sulfo-, cyano-, or nitro-, substituted phenyl; or electron-withdrawing substituted phenylethyl. More preferably, R4 is methyl, β-cyanoethyl, or 4- nitrophenylethyl. Most preferably, R4 is hydrogen, methyl, or β-cyanoethyl. Electron-withdrawing substituents are typically halo, cyano, nitro, sulfo, or mono-, di-, or trihalomethyl, and the like. Halogen atom substituents are usually fluoro, chloro, bromo, or iodo; and preferably, they are fluoro or chloro. "Electron-withdrawing" denotes the tendancy of a substituent to attract valence electrons of the molecule of which it is apart, i.e. it is electronegative, e.g. March, Advanced Organic Chemistry, pgs. 16-18 (John Wiley, New York, 1985). For convenience, nucleotide phosphoramidates are sometimes indicated herein by a subscripted "np" or "pn" for N3'→P5' phosphoramidates and P3'→N5' phosphoramidates, respectively. Thus, "Unp U" is a dinucleotide in which a 3'-aminouridine and a deoxyuridine are linked by an N3'→P5' phosphoramidate linkage. Similarly, 2'-fluoro substituents are indicated by a superscripted "f". Thus, "Uf np U" is a dinucleotide in which the 3'-most 2'-fluorouridine is linked to a uridine by an N3'→P5' phosphoramidate linkage. A single leading subscripted "p" indicates to a 5' monophosphate, and a single trailing subscripted "n" indicates a 3'-amino.
As used herein, "nucleoside" includes the natural nucleosides, including 2'-deoxy and 2'-hydroxyl forms, e.g. as described in Kornberg and Baker, DNA Replication, 2nd Ed. (Freeman, San Francisco, 1992). "Analogs" in reference to nucleosides includes synthetic nucleosides having modified base moieties and/or modified sugar moieties, e.g. described generally by Scheit, Nucleotide Analogs (John Wiley, New York, 1980). Such analogs include synthetic nucleosides designed to enhance binding properties, e.g. stability, specificity, or the like, such as disclosed by Uhlmann and Peyman (cited above).
As used herein, "pyrimidine" means the pyrimidine occurring in natural nucleosides, including cytosine, thymine, and uracil, and common analogs thereof, such as those containing oxy, methyl, methoxy, hydroxyl, amino, thio, and like, substituents. The term as used herein further includes pyrimidines with common protection groups attached, such as 4N-benzoylcytosine. Further common pyrimidine protection groups are disclosed by Beaucage and Iyer, Tetrahedron, 48:2223-23 11 (1992).
As used herein, "purine" means the purines occurring in natural nucleosides, including adenine, guanine, and hypoxanthine, and common analogs thereof, such as those containing oxy, methyl, methoxy, hydroxyl, amino, thio, and like, substituents. The term as used herein further includes purines with common protection groups attached, such as 2N-benzoylguanine, 2N-isobutrylguanine, 6N-benzoyladenine, and the like. Further common purine protection groups are disclosed by Beaucage and Iyer (cited above).
DETAILED DESCRIPTION OF THE INVENTION
The invention includes polymers of 2'-fluoro-3'-aminonucleosides, that is, oligo-2'-fluoronucleotide N3'→P5' phosphoramidates, monomers and methods for solid phase assembly of such polymers, and uses of the polymers. The invention also includes compositions comprising Watson-Crick duplexes of RNA or DNA and oligo-2'-fluoronucleotide N3'→P5' phosphoramidates and compositions comprising triplexes of DNA duplexes and oligo-2'-fluoronucleotide N3'→P5' phosphoramidates, and methods and kits for using oligo-2'-fluoronucleotide N3'→P5' phosphoramidates as DNA capture agents based on either Watson-Crick basepairing with single stranded target nucleic acids or Hoogsteen or reverse Hoogsteen triplet formation with double stranded target nucleic acids.
Preferably, oligo-2'-fluoronucleotide N3'→P5' phosphoramidates of the invention are between 2 and 30 nucleotides in length. More preferably, they are between 8 and 25 nucleotides in length; and most preferably, they are between 8 and 20 nucleotides in length.
Oligomers of the invention may be synthesized by several different approaches. In a first approach, coupling is achieved by carbon tetrachloride-driven oxidative phosphorylation of a nucleoside 3 '-amine by a 5'-H-phosphonate of the terminal nucleotide of a growing chain anchored to a solid phase support, as described, for example, by Gryaznov et al, J. Am. Chem. Soc. 116:3143-3144 (1994); and Chen et al, Nucleic Acids Research, 23: 2661-2668 (1995); which references are incorporated by reference. Monomers for this synthetic approach have the following form: ##STR2## where B is defined as above and R1 is hydrogen or a hydroxyl protecting group, such as triphenylmethyl (i.e., trityl), p-anisyldiphenylmethyl (i.e., monomethoxytrityl or MMT), di-p-anisylphenylmethyl (i.e., dimethoxytrityl or DMT), pivaloyl, acetyl, 4-methoxytetrahydropyran-4-yl, tetrahydropyranyl, phenoxyacetyl, isobutyloxycarbonyl, pixyl, benzyl, trialkylsilyl having from 3 to 9 carbon atoms, 9-fluorenylmethyl carbamate (Fmoc), or the like. Preferably, R1 is DMT. Greene and Wuts, Protective Groups in Organic Synthesis, 2nd Edition (John Wiley, New York, 1991) provides extensive guidance on the selection of protecting groups for the various embodiments of the invention.
A general scheme for preparing the above monomers is shown in FIG. 1. Briefly, a ribonucleoside is transformed into a 5'-hydroxyl-protected-2',3'-anhydroxylnucleoside, after which the 2',3'-epoxy ring is opened by treatment with sodium azide, or like reagent, to form a 5'-hydroxyl-protected-3'-azido-3'-deoxyarabinonucleoside. After purification, the 5'-hydroxyl-protected-3'-azido-3'-deoxyarabinonucleoside is fluorinated at the 2' position by treatment with diethylaminosulfur trifluoride (DAST), or like reagent, after which the azido group is reduced to give the above monomer. The monomers are then employed for chain assembly essentially as described by Chen et al and Gryaznov et al (cited above).
In a second approach, oligomers of the invention are synthesized via an exchange reaction between a phosphoramidite group on a monomer and a 3'-amino of an anchored chain, as illustrated by Scheme 4 in FIG. 4. Monomers for this synthetic approach have the following form: ##STR3## where B and R1 are defined as above, R5 is an amino protecting group, and R' and R" taken together with the nitrogen form an alkyl- or arylamino leaving group. More particularly, R' and R" taken separately each are alkyl, aralkyl, cycloalkyl, and cycloalkylalkyl containing up to 10 carbon atoms in total. Preferably R' and R" taken separately are alkyl having from 1 to 5 carbon atoms. Most preferably, R' and R" taken separately are isopropyl. R' and R" taken together form an alkylene chain containing up to 5 carbon atoms in the principal chain and a total of up to 10 carbon atoms with both terminal valence bonds of said chain being attached to the nitrogen atom to which R' and R" are attached; or R' and R" when taken together with the nitrogen atom to which they are attached form a saturated nitrogen heterocycle which may contain one or more additional heteroatoms from the group consisting of nitrogen, oxygen, and sulfur. More preferably, R' and R" taken together and with the nitrogen to which they are attached are pyrrolidino, morpholino, or piperidino. Most preferably, R' and R" taken together and with the nitrogen to which they are attached are morpholino. Preferably, R5 is trityl, and more preferably, R5 is 4-methoxytrityl.
Phosphoramidite monomers of the invention are prepared as follows, as illustrated in FIGS. 2 and 3. As above, a ribonucleoside is transformed into a 5'-hydroxyl-protected-2',3'-anhydroxylnucleoside, after which the 2',3'-epoxy ring is opened by treatment with sodium azide, or like reagent, to form a 5'-hydroxyl-protected-3'-azido-3'-deoxyarabinonucleoside. After purification, the 5'-hydroxyl-protected-3'-azido-3'-deoxyarabinonucleoside is fluorinated at the 2' position by treatment with diethylaminosulfur trifluoride (DAST), or like reagent, after which the azido group is reduced to give a 3'-amino. After suitably protecting the 3'-amino and releasing the 5'-hydroxyl protecting group, the nucleoside is phosphitylated at the 5' oxygen to give the crude phorphoramidite monomer.
The phosphoramidite monomers are used in the following synthetic cycle:
1) detritylation with acid of the 3'-amino group of nucleoside attached to a solid support through 5'-terminus;
2) tetrazole-catalyzed amidite transfer reaction between the phosphoramidite monomer and the 3'-amino group of the nucleoside on a solid support, resulting in formation of an internucleoside phosphoramidite diester group; this may be repeated with intermediate washing with acetonitrile, or like solvent, to achieve slightly higher efficiency of chain elongation;
3) oxidation of the newly formed internucleoside phosphoramidite diester into a phosphoramidate diester group with aqueous iodine, or like oxidizing agent;
4) capping of unreacted 3'-amino groups with acetic anhydride, or like capping agent.
This cycle can be repeated, resulting in oligo-2'-fluoronucleotide N3'→P5' phosphoramidates after cleavage from the solid support and deprotection with ammonia.
A wide variety of solid phase supports may be used with the invention, including microparticles made of controlled pore glass (CPG), highly cross-linked polystyrene, acrylic copolymers, cellulose, nylon, dextran, latex, polyacrolein, and the like, disclosed in the following exemplary references: Meth. Enzymol., Section A, pages 11-147, vol. 44 (Academic Press, New York, 1976); U.S. Pat. Nos. 4,678,814; 4,413,070; and 4,046;720; and Pon, Chapter 19, in Agrawal, editor, Methods in Molecular Biology, Vol. 20, (Humana Press, Totowa, N.J., 1993). Supports further include commercially available nucleoside-derivatized CPG and polystyrene beads (e.g. available from Applied Biosystems, Foster City, Calif.); polystyrene grafted with polyethylene glycol (e.g., TentaGel™, Rapp Polymere, Tubingen Germany); and the like. Selection of the support characteristics, such as material, porosity, size, shape, and the like, and the type of linking moiety employed depends on a variety of factors, such as protection groups employed, length of final product, quantity of final product, and the like. Exemplary linking moieties are disclosed in Pon et at, Biotechniques, 6:768-775 (1988); Webb, U.S. Pat. No. 4,659,774; Barany et al, International patent application PCTFLIS91/06103; Brown et al, J. Chem. Soc. Commun., 1989: 891-893; Damha et at, Nucleic Acids Research, 18:3813-3821 (1990); Beattie et al, Clinical Chemistry, 39:719-722 (1993); Maskos and Southern, Nucleic Acids Research, 20: 1679-1684 (1992); and the like.
Preferably, thermal stability of oligo-2'-fluoronucleotide N3'→P5' phosphoramidates are determined by way of melting, or strand dissociation, curves. The temperature of fifty percent strand dissociation is taken as the melting temperature, Tm, which, in turn, provides a convenient measure of stability. Tm measurements are typically carried out in a saline solution at neutral pH with target and oligomer concentrations at between about 1.0-3.0 μM. Typical conditions are as follows: 150 mM NaCl and 10 mM MgCl2 in a 10 mM sodium phosphate buffer (pH 7.0) or in a 10 mM Tris-HCl buffer (pH 7.0); or like conditions. Data for melting curves are accumulated by heating a sample, e.g. a duplex or triplex, of the oligo-2'-fluoronucleotide N3'→P5' phosphoramidate and its complementary single stranded or double stranded oligonucleotide from 10-15° C. to about 85°-90° C. As the temperature of the sample increases, absorbance of 260 nm light is monitored at 1° C. intervals, e.g. using a Cary (Australia) model 1E or a Hewlett-Packard (Palo Alto, Calif.) model HP 8459 UVNIS spectrophotometer and model HP 89100A temperature controller, or like instruments.
In embodiments where triplex formation is desired, there are constraints on the selection of complementary sequences. Generally, third strand association via Hoogsteen type of binding is most stable along homopyrimidine-homopufine tracks in a double stranded target. Usually, base triplets form in T-A*T or C-G*C motifs (where "-" indicates Watson-Crick pairing and "*" indicates Hoogsteen type of binding); however, other motifs are also possible. For example, Hoogsteen base pairing permits parallel and antiparallel orientations between the third strand (the Hoogsteen strand) and the purine-rich strand of the duplex to which the third strand binds, depending on conditions and the composition of the strands. There is extensive guidance in the literature for selecting appropriate sequences, orientation, conditions, nucleoside type (e.g. whether ribose or deoxyribose nucleosides are employed), base modifications (e.g. methylated cytosine, and the like) in order to maximize, or otherwise regulate, triplex stability as desired in particular embodiments, e.g. Roberts et al, Proc. Natl. Acad. Sci., 88:9397-9401 (1991); Roberts et at, Science, 258:1463-1466 (1992); Distefano et al, Proc. Natl. Acad. Sci., 90:1179-1183 (1993); Mergny et at, Biochemistry, 30:9791-9798 (1991); Cheng et at, I. Am. Chem. Soc., 114:4465-4474 (1992); Beat and Dervan, Nucleic Acids Research, 20:2773-2776 (1992); Beat and Dervan, J. Am. Chem. Soc., 114:4976-4982 (1992); Giovannangeli et at, Proc. Natl. Acad. Sci., 89:8631-8635 (1992); Moser and Dervan, Science, 238:645-650 (1987); McShan et at, J. Biol. Chem., 267:5712-5721 (1992); Yoon et at, Proc. Natl. Acad. Sci., 89:3840-3844 (1992); Blume et al, Nucleic Acids Research, 20:1777-1784 (1992); and the like. Generally, after one of the oligonucleotide moieties forms a Watson-Crick duplex with a pyrimidine-rich or purine-rich track in a target polynucleotide, the remaining oligonucleotide components bind to the major groove of the duplex to form a triplex structure.
Compounds of the invention can be employed as diagnostic probes to detect the presence of one or more target polynueleotides in a wide range of samples, including environmental samples, e.g. from public water supplies, samples from foodstuffs, and from other biological samples, such as blood, saliva, semen, amniotic fluid, tissue homogenates of plants or animals, or of human patients, and the like. The use of nucleic acid probes in human diagnostics, forensics, and genetic analysis has been extensively reviewed. For example, the following references describe many diagnostic applications of nucleic acid probes for which the present invention can be usefully employed: Caskey, Science 236:1223-1228 (1987); Landegren et al, Science, 242:229-237 (1988); and Arnheim et al, Ann. Rev. Biochem., 61:131-156 (1992). Moreover, there is extensive guidance in the literature concerning the selection of hybridization conditions, labeling means, and the like, which is applicable to the practice of the present invention, e.g. Wetmer, Critical Reviews in Biochemistry and Molecular Biology 26:227-259 (1991); and the like.
Oligo-2'-fluoronucleoside N3'→P5' phosphoramidates of the invention may be used in essentially any of the known solution or solid phase hybridization formats, such as those in which the analyte is bound directly to a solid phase, or sandwich hybridizations in which the analyte is bound to an oligonucleotide that is, in turn, bound to a solid phase. Oligo-2'-fluoronucleoside N3'→P5' phosphoramidates are particularly useful in conjunction with branched polymer signal amplification schemes, such as those disclosed by Urdea et al, U.S. Pat. No. 5,124,246; Wang et al, U.S. Pat. No. 4,925,785; and the like. Urdea et al and Wang et al are incorporated by reference for their description of such hybridization assays. In such embodiments, the oligo-2'-fluoronucleoside N3'→P5' phosphoramidate preferably serves as a highly stable and specific "capture" probe by binding to a target polynucleotide analyte of interest.
Kits incorporating oligo-2'-fluoronucleotide N3'→P5' phosphoramidates can take a variety of forms depending on the particular embodiment, the type of assay format employed, and the labeling scheme employed. Generally, kits of the invention comprise an oligo-2'-fluoronucleoside N3'→P5' phosphoramidate specific for a given target polynucleotide, a hybridization buffer, and a signal generation moiety. Kits of the invention may further comprise wash buffers for removing unbound label and/or oligo-2'-fluoronucleoside N3'→P5' phosphoramidate, solid phase supports such as derivatized magnetic beads, or the like; and prehybridization buffers containing blocking agents, e.g. Denhardt's solution, sonicated salmon sperm DNA, detergents such as 1% SDS, or the like, for minimizing nonspecific binding of the oligo-2'-fluoronucleoside N3'→P5' phosphoramidate or other nucleosidic binding components, such as amplifier strands. An exemplary hybridization buffer comprises the following reagents: 100-150 mM NaCl, 10 mM MgCl2, and 10 mM Tris-HCl (pH 7.0).
Compounds of the invention may be used as DNA capture agents by forming highly stable triplex between a double stranded target DNA and an oligo-2'-fluoronucleotide N3'→P5' phosphoramidate of a predetermined sequence. This application is particularly useful in manipulating multiple double stranded DNAs such as DNA sequencing plasmids of PCR amplicons that contain complementary sequences for binding via triplex formation with the oligo-2'-fluoronucleotide N3'→P5' phosphoramidates, such as described ordinary oligonucleotides by Cantor et at, U.S. Pat. No. 5,482,836, which patent is incorporated by reference. Preferably, compounds of the invention are biotinylated or covalently attached to a like binding moiety, such as a hapten. After contacting with target DNAs in a sample under conditions that permit triplex formation, the target DNAs are extracted from the sample by contacting the biotin, or like binding moiety, with its receptor which preferably is attached to a solid support, such as a magnetic bead. After washing, the captured DNAs may be release under conditions that destroy the stability of the triplex structure, e.g. mild alkaline buffer, pH 8-10.
Signal generation moieties are molecular structures that directly or indirectly generate a signal, e.g. fluorescent, colorimetric, radioactive, or the like, that can be detected by conventional means. Direct signal generation means that the moiety producing a signal is covalently linked to the oligo-2'-fluoronucleoside N3'→P5' phosphoramidate, e.g. as with the covalent attachment of a fluorescent dye, enzyme, or the like. Indirect signal generation means that a particular moiety, e.g. an oligonucleotide tail conjugated to a oligo-2'-fluoronucleoside N3'→P5' phosphoramidate, is one component of a multi-component system that produces a signal. Preferably, the signal generation moieties comprises an oligonucleotide tail of about 12 to about 50 nucleotides in length. In one aspect of this preferred embodiment, a signal is generated indirectly by providing a second oligonucleotide which is complementary to the tail and which has a fluorescent dye covalently attached. Attaching fluorescent dyes to oligonucleotides is well known in the art, e.g. U.S. Pat. Nos. 4,997,828; 5,151,507; 4,855,225; 5,188,934; Eckstein, editor (cited above); and the like.
Compounds of the invention can be employed as antisense or anti-gene compounds for treating disease. The components included in such pharmaceutical compositions of the invention depend on several factors, including the nature of the disease or condition being treated, the location of disease lesions, the mode of drug delivery and/or administration contemplated, the latter of which can include in vivo administration by way of regional or systemic perfusion, topical application, intranasal administration, administration by implanted or transdermal sustained release systems, and the like, as well as ex vivo administration for use in bone marrow purging. A preferred method of administration of oligonucleotides comprises either regional or systemic perfusion. According to a method of regional perfusion, the afferent and efferent vessels supplying the extremity containing a lesion, e.g. a cancerous lesion, are isolated and connected to a low-flow perfusion pump in continuity with an oxygenator and heat exchanger. The iliac vessels may be used for perfusion of the lower extremities. The axillary vessels are cannulated high in the axilla for upper extremity lesions. A pharmaceutical composition containing an oligo-2'-fluoronucleotide N3'→P5' phosphoramidate is added to the perfusion circuit, and the perfusion is continued for an appropriate time period, e.g. an hour. Perfusion rates of from 100 to 150 ml/minute may be employed for lower extremity lesions, while half that rate should be employed for upper extremity lesions. Systemic heparinization may be used throughout the perfusion, and reversed after the perfusion is complete. This isolation perfusion technique permits administration of higher dosed of chemotherapeutic agent than would otherwise be tolerated upon infusion into the arterial or venous systemic circulation.
Generally a pharmaceutical composition of the invention facilitates the delivery of an effective amount of the active drug to a desired site in a manner consistent with patient safety and comfort. An effective amount of an oligo-2'-fluoronucleotide N3'→P5' phosphoramidate depends on several factors, including the disease or condition being treated, the method of administration, the scheduling of the administration, the condition of the patient, and the like. Typically, a parentially administered dose will be in the range of about 1 g/kg/day to about 100 mg/kg/day of patient body weight. A key factor in selecting an appropriate dose for a given condition or disease is the therapeutic result, as measure by standard criteria well known to the medical practitioner, e.g. for oncological applications see: Cancer: Principles and Practice of Oncology, 3rd Edition, edited by V. T. DeVita et al (Lippincott Company, Philadelphia, 1989).
Pharmaceutical compositions of the invention include a pharmaceutical carrier that may contain a variety of components that provide a variety of functions, including regulation of drug concentration, regulation of solubility, chemical stabilization, regulation of viscosity, absorption enhancement, regulation of pH, and the like. For example, in water soluble formulations the pharmaceutical composition preferably includes a buffer such as a phosphate buffer, or other organic acid salt, preferably at a pH of between about 7 and 8. For formulations containing weakly soluble oligonucleotides, microemulsions may be employed, for example by using a nonionic surfactant such as Tween 80 in an amount of 0.04-0.05% (w/v), to increase solubility. Other components may include antioxidants, such as ascorbic acid, hydrophilic polymers, such as, monosaccharides, disaccharides, and other carbohydrates including cellulose or its derivatives, dextrins, chelating agents, such as EDTA, and like components well known to those in the pharmaceutical sciences, e.g. Remington's Pharmaceutical Science, latest edition (Mack Publishing Company, Easton, Pa.).
Compounds of the invention include the pharmaceutically acceptable salts thereof, including those of alkaline earths, e.g. sodium or magnesium, ammonium or NX4 +, wherein X is C 1-4 alkyl. Other pharmaceutically acceptable salts include organic carboxylic acids such as acetic, lactic, tartaric, malic, isethionic, lactobionic, and succinic acids; organic sulfonic acids such as methanesulfonic, ethanesulfonic, and benzenesulfonic; and inorganic acids such as hydrochloric, sulfuric, phosphoric, and sulfamic acids. Pharmaceutically acceptable salts of a compound having a hydroxyl group include the anion of such compound in combination with a suitable cation such as Na+, NH4 +, or the like.
Sustained release systems suitable for use with the pharmaceutical compositons of the invention include semi-permeable polymer matrices in the form of films, microcapsules, or the like, comprising polylactides, copolymers of L-glutamic acid and gamma-ethyl-L-glutamate, poly(2-hydroxyethyl methacrylate), and like materials. Sustained release systems also include liposomally entrapped oligonucleotides, e.g. as described in Liposome Technology, Vol. II, Incorporation of Drugs, Proteins, and Genetic Material (CRC Press).
Experimental Methods
Phosphodiester oligodeoxyribonucleotides and oligoribonucleotides were prepared on an ABI 380B DNA synthesizer using standard protocol via the phosphoramidite method.18 Oligonucleotide N3'→P5' phosphoramidates, containing 2'-deoxy- and one or two 2'-fluoronucleosides were synthesized using the oxidative phosphorylation method on a ABI 394 synthesizer as previously described.4c Uniformly modified oligo-2'-fluoronucleotide N3'→P5' phosphoramidates were prepared by amidite transfer reaction on an ABI 380B synthesizer using the following protocol:
1) detritylation, 5% dichloroacetic acid in dichloromethane, 1 min
2) coupling, 0.1M phosphoramidite 2 and 0.45M tetrazole in acetonitrile, 3 min
3) oxidation, 0.1M iodine in tetrahydrofuran/pyridine/water, 10/10/1, v/v/v, 1 min
4) capping, acetylation of unreacted 3'-amino groups by standard ABI capping solutions, 30 sec
Chemical steps within the cycle were followed by acetonitrile washings and flushings with dry argon for 0.2-0.4 min. After cleavage from the solid support and deprotection with concentrated aqueous ammonia, 1-1.5 h, 55° C., oligonucleotides were analyzed and purified by IE HPLC. Oligonucleotides were desalted on Pharmacia NAP-5 or NAP-10 gel filtration columns immediately after purification and stored frozen or lyophilized at -18° C.
Preparation of the 5'-phosphorylated oligonucleotides was done upon sulfone-derivatized CPG,19 and 5'-hydroxyl oligomers were synthesized upon oligonucleotide-succinyl CPG.
Oligonucleotide thermal dissociation experiments were carded out as previously described,4b with melting buffers as listed in Table 1.
Dionex DX300 or DX500 systems were used for IE analysis and purification of oligonucleotides. A Pharmacia MonoQ 10/10 column was used for analysis and purification of crude oligomers, eluted with a 2% per minute gradient of 1.5M NaCl in 10 mM NaOH. A Dionex NucleoPac PA100 column, eluted with a 1.5% per minute gradient of 1.5M NaCl in 10 mM NaOH was used for all other IE HPLC analysis. A Hewlett Packard Hypersil ODS, 5 g column on a Waters HPLC system was used for RP HPLC, with a 1% per minute gradient of acetonitrile in 0.1M triethylammonium acetate, pH 7.0.
NMR spectra were recorded on a Bruker DRX-400 spectrometer. Chemical shifts are reported relative to TMS, CCl3 F, and H3 PO4, for 1 H, 19 F, and 31 P spectra, respectively.
Thin layer chromatography (TLC) was performed on Whatman polyester-backed silica gel plates with methanol/dichloromethane eluents.
Acid hydrolysis of 0.17 OD260 of the dimer dUf np T was done in 25 gL of 64% acetic acid, 2 h at 55° C., and the reaction mixture was analyzed by RP HPLC. Approximately 83% of the dimer, retention time (Rt) 15.0 min, was hydrolyzed to mainly 5'-thymidylic acid, Rt 10.6 min, and 2'-fluor-3'-aminouridine, Rt 11.2 min, as were identified by co-injection with authentic standards. Also, ˜7.5% of thymidine, Rt 12.1 min, was found in the reaction mixture.
Acid hydrolysis of the oligonucleotide phosphoramidates (Table 2), 1-30D260 of each, was carded out at room temperature in 0.1-0.2 mL of 10% acetic acid, pH 2.2 or in sodium acetate buffers, pH 4.7 and 5.3. For enzymatic digestion, 0.2 OD260 of oligonucleotides 19 and 22 (Table 1) were treated with 0.02 units of snake venom phosphodiesterase and 0.8 units of alkaline phosphatase, both from Sigma, in 0.2 mL of Tris-HCl buffer, pH 8.9, at room temperature. Aliquots from the reaction mixtures were taken at multiple time points and analyzed by IE HPLC.
EXAMPLE 1
5'-DMT-O-2'-fluoro-3 '-aminouridine
Compound 1, 5'-DMT-2'-fluoro-3'-aminouridine was prepared according to Scheme 1 shown in FIG. 1. This monomer was used for incorporation of 2'-fluoro-3'-aminouridine into oligonucleotide phosphoramidates by the oxidative phosphorylation method.4 First, uridine 3 was transformed into the 5'-DMT-2',3'-anhydrolyxouridine 4 by successive, one pot reaction with DMT-chloride, mesyl chloride, and sodium hydroxide. The 2',3'-epoxy ring was then opened by treatment with sodium azide,10 producing 5'-DMT-3'-azido-3'-deoxyarabinouridine 5 as the main product, and isomeric 5'-DMT-2'-azido-2'-deoxyxylouridine as a by-product, in approximately a 2:1 ratio. Compound 5 was isolated by silica gel chromatography and then fluorinated with diethylaminosulfur trifluoride (DAST) to give 6. Finally, the azido group of 6 was reduced with hydrogen over palladium catalyst, giving 5'-DMT-2'-fluoro-3'-aminouridine 1. The structure of nucleoside 1 was confirmed by 1 H and 19 F NMR analysis and by mass spectrometry (FIG. 5). More particularly, the steps were carded out as follows:
5'-0-DMT-2',3'-anhydrolyxouridine (compound 4, FIG. 1) was prepared as follows: To 10.0 g (41 mmol) of dry 3 in 250 mL anhydrous pyridine was added 14.6 g (43 mmol) of dimethoxytrityl chloride. The mixture was stirred overnight, then cooled on ice and 8.0 mL (103 mmol) methanesulfonyl chloride was added. After stirring at room temperature for 4 h, the reaction was quenched with 1 mL H2 O and the solvent removed in vacuo. The residue was suspended in 500 mL CH2 Cl2, washed with water (3×250 mL), and reconcentrated in vacuo to a foam. The foam was dissolved in 450 mL dioxane and 250 mL of 1N NaOH added. After stirring for 4 h, the dioxane was removed in vacuo, and the resultant slurry extracted with 500 mL of CH2 Cl2. Concentration of the CH2 Cl2 layer and flash chromatography provided 13.8 g (64%) of product as a light yellow powder. Mass-spectrometry, FAB+, M+H+, calculated: 529.1975, observed: 529.1963. 1 H NMR δ7.58 (d, J=8.2 Hz, 1H), 7.5-7.2 (mm, 10H), 6.86 (d, J=8.2 Hz, 4H), 6.20 (s, 1 H), 5.68 (d, J=8.1 Hz, 1H), 4.20 (dd, J=5.8, 5.8 Hz, 1H), 3.96 (d, J=2.9 Hz, 1H), 3.92 (d, J=3.0 Hz, 1H), 3.82 (s, 6H), 3.47 (dd, J=5.9, 9.7 Hz, 1H), 3.38 (dd, I =5.7, 9.6 Hz, 1H).
5'-O-DMT-3'-azido-3'-deoxyarabinouridine (compound 5, FIG. 1) was prepared as follows: To 13.8 g (26 mmol) of 4 in 500 mL acetone was added 200 mL H2 O and 12.0 g (185 mmol) NAN3. The mixture was refluxed overnight, then concentrated/n vacuo to remove the acetone. The resultant slurry was extracted with 600 mL CH2 Cl2, which was in turn was washed with water (3×250 mL). Concentration of the CH2 Cl2 layer and flash chromatography of the crude product provided 6.0 g (40%) of a pale yellow solid. Mass-spectrometry, FAB+, M+H+, calculated: 572.2145, observed: 572.2147. 1 H NMR δ 9.4 (br s, 1H), 8.07 (d, J=8.1 Hz, 1H), 7.4-7.3 (mm), 6.89 (d, J=7.9 Hz, 4H), 6.10 (d, J=5.5 Hz, 1H), 5.46 (d, J=8.1 Hz, 1H). 4.55 (m, 1H), 4.19 (dd, J=7.2, 7.7 Hz, 1H), 3.84 (m, 1H), 3.83 (s, 6H), 3.64 (dd J=2.6, 11.3 Hz, 1H), 3.43 (dd, J=2.6, 11.4 Hz, 1H).
5'-O-DMT-2'-fluoro-3'-azido-2',3'-dideoxyuridine (compound 6, FIG. 1) was prepared as follows: To 6.0 g (10.5 mmol) of 5 in 120 mL anhydrous DMF was added 2.4 mL (18.2 mmol) of diethylaminosulfur trifluoride. The mixture was stirred for 16 h, then poured into 300 mL of cold saturated aqueous NaHCO3. The product was extracted with 500 mL ethyl acetate, which in turn was washed with water (2×500 mL). Concentration of the organic layer and flash chromatography of the crude product provided 2.9 g (48%) of an off-white solid. Mass-spectrometry, FAB+, M+·, calculated: 573.2024, observed: 573.2011. 1 H NMR δ 8.95 (br s, 1H), 7.91 (d, J=8.1 Hz, 1H), 7.2-7.4 (mm, 11H), 6.88 (d, J=8.6 Hz, 4H), 6.02 (d, J=17.2 Hz, 1H), 5.42 (d, J=8.1 Hz, 1H), 5.27 (dd, J=3.7, 55.0 Hz, 1H), 4.24 (m, 1H), ˜4.21 (partially overlapping with signal 4.24 ppm, presumed ddd, J=4, 4, -25 Hz, 1H), 3.82 (s, 6H), 3.71 (d, J=11.4 Hz, 1H), 3.49 (d, J=11.3 Hz, 1H); 19 F NMR δ -196.7 (dddd, est. J=3, 17, 25, 55 Hz).
5'-O-DMT-2'-fluoro-3'-amino-2',3'-dideoxyuridine (compound 6, FIG. 1) was prepared as follows: To 2.9 g (5.1 mmol) of 6 in 75 mL of 95% ethanol was added 0.5 g of 10% palladium on carbon. The mixture was hydrogenated at 40 psi overnight and then the catalyst removed by filtration. The solvent was removed in vacuo, and the resultant solid purified by flash chromatography to give 1.2 g (43%) of product as a white powder. Mass-spectrometry, FAB+, M+H+, calculated: 548.2197, observed: 548.2206. 1 H NMR δ 8.04 (d, J=8.0 Hz, 1H), 7.9 (br m, 1H), 7.2-7.4 (mm), 6.85 (d, J=8.4 Hz, 4H), 6.00 (d, J=16.2 Hz, 1H), 5.32 (d, J=8.7 Hz, 1H), 4.83 (dd, J=3.9, 52.2 Hz, 1H), 3.88 (br d, J=11 Hz, 1H), 3.8 (s, 6H), 3.8-3.7 (mm, 2H), 3.52 (dd, J=2.6, 11.1 Hz, 1H); 19 F NMR δ -200.1 (ddd, J=16.4, 27.5, 52.1 Hz).
EXAMPLE 2
N4 -benzoyl-2'-fluoro-3'-(4-methoxytrityl)amino-2',3 '-dideoxyuridine 5'-(2-cyanoethyl N,N-(diisopropyl)phosphoramidite
Oligomers of the invention were also synthesized from phosphoramidite monomer, as illustrated by compound 2u of Scheme 2. Uridine 3 was mesylated and then selectively benzoylated with accompanying formation of the 2,2'-anhydrocycle by treatment with sodium benzoate according to literature procedure.11 These reactions resulted in compound 7 with 69-77% overall yields. By another literature method,12 2,3'-anhydroarabinonueleoside 7 was transformed into 2',3'-anhydrolyxoufidine 8 in two steps. This involved treatment of 7 with hydrochloric acid to form 3 '-mesyl-5 '-benzoylarabinouridine, which upon treatment with ammonium hydroxide closed to form the lyxo-2',3'-epoxide 8 in 63-77% overall yields. Then, also according to published procedure,13 2',3'-anhydrolyxonucleoside 8 was heated with ammonium azide. Contrary to literature suggestion, this reaction was not completely stereoselective, but produced a chromatographically unresolvable mixture of the desired 5'-benzoyl-3'-azidoarabinonucleoside 9 and it's 2'-azido-2'-deoxyregioisomer 9i in approximately a 2.5:1 ratio. Crude arabinonucleoside 9 was fluorinated with DAST to give 2'-fluoro-3'-azidonucleoside 10, then catalytically hydrogenated to give 2'-fluoro-3'-aminonucleoside 11, which was separable from its regioisomer by silica gel chromatography. Protection of the 3'-amine with a monomethoxytrityl (MMT) group, followed by 5'- debenzoylation produced intermediate 13, with 5'-phosphitylation producing the desired phosphoramidite building block 2u in a 22% overall yield from anhydronucleoside 8. More particularly, the steps were carried out as follows:
3'-O-Methanesulfonyl-5'-O-benzoyl-2,2'-anhydroarabinouridine (compound 7, FIG. 2) was prepared in two steps from 3 (FIG. 2) according to the procedure of Codington et al.11 in 69-77% overall yields.
5'-O-benzoyl-2',3'-anhydrolyxouridine (compound 8, FIG. 2) was prepared in two steps from 7 according to the procedure of Codington et al.12 in 63-77% overall yields.
3'-azido-5'-O-benzoyl-3'-deoxyarabinouridine (compound 9, FIG. 2) was prepared from 8 and anhydrous NH4 N3,20 according to the procedure of Reichman et al.,13 but without successful recrystallization. Mass yields were 98% or greater, but NMR suggested 25-35% of the regioisomer, 2'-azido-5'-O-benzoyl-2'-deoxyxylouridine, 9i, which coeluted with the desired product by silica gel TLC. 1 H NMR, Major component, 9: δ 10.8 (br s, 1H), 8.11 (d, J=7.5 Hz, 2H), 7.68 (d, J=8.1 Hz, 1H), 7.62 (d, J=7.3 Hz, 1H), 7.5 (m, 2H), 6.19 (d, J=3.6 Hz, 1H), 5.40, (d, J=8.0 Hz, 1H), 4.84 (m, 1H), 4.73 (d, J=5.7 Hz, 1H), 4.63 (br d, J=4.2 Hz, 1H), 4.2 (mm, 2H); Minor component, 9i: δ 10.6 (br s, 1H), 8.11 (d, J=7.5 Hz, 2H), 7.81 (d, J=8.1 Hz, 1H), 7.64 (d, J=7.5 Hz, 1H), 7.5 (m, 2H), 5.85 (s, 1H), 5.47, (d, J=8.1 Hz, 1H), 4.86 (m, 1H), 4.76 (d, J=5.4 Hz, 1H), 4.62 (br d, J=4.0 Hz, 1H), 4.3-4.2 (mm, 2H).
2'-fluoro-3'-azido-5'-O-benzoyl-2',3'-dideoxyuridine (compound 10, FIG. 2) was prepared as follows: To 5.0 g (13.4 mmol) of crude 9 (containing 25% 9i) in 30 mL anhydrous CH2 C12 was added 8.8 mL (66.6 mmol) of diethylaminosulfur trifluoride. After stirring for 48 h, the mixture was diluted with 100 mL CH2 Cl2 and poured into 200 mL saturated aqueous NaHCO3. When evolution of gas ceased, the CH2 Cl2 layer was washed with 100 mL fresh NaHCO3 solution and then with water (2×100 mL). Concentration of the CH2 Cl2 layer in vacuo and flash chromatography gave 3.5 g (70%) of product containing 20% of the largely chromatographically unresolvable isomeric impurity, 10i. 1 H NMR, Major component, 10: δ 8.7 (br s, 1H), 8.07 (d, J=7.4 Hz, 2H), 7.62 (d, J=7.5 Hz, 1H), 7.49 (dd, J=7.6, 7.6 Hz, 2H), 7.39 (d, J=8.1 Hz, 1H), 5.70 (d, J=21.1 Hz, 1H), 5.65 (d, J=8.2 Hz, 1H), 5.48 (dd, J=4.7, 52.9 Hz, 1H), 4.7-4.4 (unresolved), 4.32 (dd, J=4.7, 9.5 Hz, 1H), 4.27 (dd, J=4.7, 9.5 Hz, 1H); Minor component, 10i: 5 8.7 (br s, 1H), 8.03 (d, J=7.2 Hz, 2H), 7.64 (d, J=7.6 Hz, 1H), 7.51 (dd, J=7.4, 7.7 Hz, 2H), 7.33 (d, J=8.2 Hz, 1H), 5.99 (d, J=6.4 Hz, 1H), 5.67 (d, J=9 Hz, 1H), 5.40 (ddd, J=2.8, 5.0, 53.4 Hz, 1H), 4.8-4.4 (unresolved), 4.10 (mm, 2H).
2'-fluoro-3'-amino-5'-O-benzoyl-2',3'-dideoxyuridine (compound 11, FIG. 2) was prepared as follows: To 3.5 g (9.3 mmol) crude 10 (20% 10i) in 200 mL 95% ethanol was added 600 mg of 10% palladium on carbon. The suspension was hydrogenated at 40 psi overnight and then the catalyst removed by filtration. The solvent was removed in vacuo, giving 2.93 g (90%) of a light yellow solid consisting of two compounds which were resolvable by TLC. Flash chromatography provided 1.96 g (60% yield) of the desired product as a pure white solid. Mass-spectrometry, FAB+, M+H+, calculated: 350.1152, observed: 350.1152. 1 H NMR δ 8.14 (br s, 1H), 8.06 (d, J=7.1 Hz, 1H), 7.64 (dd, J=7.4, 7.4 Hz, 1H), 7.57 (d, J=8.2 Hz, 1H), 7.50 (dd 7.7, 7.8 Hz, 1H), 5.86 (d, J=18.5 Hz, 1H), 5.51 (d, J=8.2 Hz, 1H) 5.00 (dd, J=4.3, 52.4 Hz, 1H), 4.81 (dd J=2.2, 12.8 Hz, 1H), 4.73 (dd, J=3.5, 12.7 Hz, 1H), 4.14 (ddd, J=2, 3, 10.2 Hz, 1H), 3.57 (ddd, J=4, 10.5, 26.6 Hz, 1H); 19 F NMR δ -198.3 (ddd, J=18.5, 26.4, 52.2 Hz).
2'-fluoro-3'-(4-methoxytrityl)amino-2',3'-dideoxyuridine (compound 12, FIG. 2) was prepared as follows: To 1.0 g (2.9 mmol) of 11 in 50 mL anhydrous pyridine was added 1.0 g (3.2 mmol) 4-methoxytrityl chloride. The mixture was stirred overnight, 5 mL saturated aqueous NaHCO3 was added, and the mixture concentrated in vacuo to an oil. The oil was dissolved in 125 mL ethyl acetate, which was washed with water (3×100 mL) and reconcentrated in vacuo to 2.05 g of foam.
The foam was dissolved in a mixture of 40 mL methanol, 40 mL dioxane, and 10 mL water. NaOH (1 g, 25 mmol) was added and the mixture stirred overnight. The solution was concentrated in vacuo to a syrup, which was dissolved in 100 mL ethyl acetate and washed with water (3×100mL). Concentration in vacuo of the organic layer gave 1.11 g of a foam, which upon flash chromatography gave 1.05 g (76%) of a white solid. Mass-spectrometry, FAB+, M+H+, calculated: 518.2091, observed: 518.2076. 1 H NMR δ 8.64 (br d J=4.2 Hz, 1H), 8.14 (br s, 1H), 7.57 (mm, 5H), 7.48 (d J=8.7 Hz, 1H), 7.3 (mm, 8H), 6.83 (d J=8.8 Hz, 2H), 5.67 (d, J=17.7 Hz, 1H), 5.62 (d, J=8.1 Hz, 1H), 4.23 (m, 2H), 4.03 (br d, J=10.2 Hz, 1H), 3.80 (s, 3H), 3.31 (dddd, J=3.6, 10.3, 10.9, 25.8 Hz, 1H), 2.80 (dd, J=3.6, 50.9 Hz, 1H), 2.51 (dd, J=3.0, 11.2 Hz, 1H); 19 F NMR δ -192.5 (dddd, J=2.9, 17.7, 26.1, 50.9 Hz).
N4 -benzoyl-2'-fluoro-3'-(4-methoxytrityl)amino-2',3'-dideoxyuridine 5'-(2-cyanoethyl N,N-diisopropyl)phosphoramidite (compound 2u, FIG. 2) was prepared as follows: To 700 mg (1.35 mmol) of 12 in 20 mL anhydrous CH2 Cl2 was added 200 mg (1.17 mmol) of diisopropylammonium tetrazolide and 0.5 mL (1.57 mmol) of 2-cyanoethyl N,N,N',N'-tetraisopropylphosphorodiamidite. After stirring the mixture for 3 h, the solvent was removed in vacuo and the residue purified on a Chromatotron, using 4 mm plates and eluting with 0-3% methanol, 0.5% triethylamine in CH2 Cl2. The product was concentrated in vacuo to an oil, which was dissolved in 10 mL CH2 Cl2 and precipitated by slow addition into 100 mL of rapidly stirred hexane. After decanting the supernatant, the product was vacuum desiccated over P2 O5, giving 680 mg (70%) of white powder. Mass-spectrometry, FAB+, M+H+, calculated: 718.3170, observed: 718.3194. 19 F NMR δ -190.9 (ddd, J=21.7, 21.8, 51.3 Hz); 31 P NMR δ 150.5, 149.5.
EXAMPLE 3
N4 -benzoyl-2'-fluoro-3'-(4-methoxytrityl)amino-2',3'-dideoxycytidine 5'-(2-cyanoethyl N,N-diisopropyl)phosphoramidite
Crude intermediate 10 was used for preparation of the appropriately protected cytidine phosphoramidite 2c (Scheme 3, FIG. 3). The uracil base of 10 was converted to cytosine by adaptation of the method of Reese et al.14 Subsequent 4-N benzoylation and reduction of the 3'-azido to an amino group gave compound 13, which was separable from its regioisomer by silica gel chromatography. Protection of the 3'-amine with an MMT group, followed by selective 5'-O-debenzoylation produced intermediate 15. Subsequent 5'-phosphitylation lead to desired phosphoramidite 2c in a 10% overall yield based on anhydronucleoside 8. More particularly, the steps were carded out as follows:
N4,5'-O-dibenzoyl-2'-fluoro-3'-amino-2',3'-dideoxycytidine (compound 13, FIG. 3) was prepared as follows: To 6.9 g (18.4 mmol) of crude 10 (containing 35% 10i) in 50 mL anhydrous CH3 CN was added an ice-cold solution of 11.7 g (169 mmol) 1,2,4-triazole and 3.35 mL (36.1 mmol) POCl3 in 90 mL anhydrous CH3 CN. The mixture was cooled in an ice bath and anhydrous triethylamine (23 mL, 165 mmol) was added, then the reaction allowed to warm to room temperature with stirring. After 90 min, 15 mL (108 mmol) triethylamine and 4 mL water were added and the mixture stirred for 10 min. The solvent was removed in vacuo, then 250 mL ethyl acetate was added, and the solution was TLC indicated a fluorescent intermediate with the same mobility as the starting material.
The mixture was concentrated in vacuo to 6.7 g of a foam. Dioxane (100 mL) and 20 mL concentrated aqueous ammonia were added, and after 3 h, the mixture was concentrated in vacuo to a yellow gel. The gel was dissolved in 100 mL ethyl acetate and washed with water (3×200 mL). Concentration in vacuo and vacuum desiccation over P2 O5 yielded 5.4 g of a solid which gave only one spot by silica gel TLC. Only two significant signals were observed by 19 F NMR, Major component: δ -192.8 (ddd, J=22.8, 22.8, 53.1 Hz); Minor component: δ -200.7 (ddd, J=13.6, 19.9, 53.4 Hz).
Anhydrous pyridine (100 mL) was added and the solution cooled to 4° C. Benzoyl chloride (11.7 mL 100 mmol) was added with stirring and the mixture allowed to warm to room temperature. After 2 h, 5 mL water was added and the solvent removed in vacuo, giving a brown oil, which was dissolved in 200 mL ethyl acetate, washed with water (3×200 mL), and then reconcentrated in vacuo to an oily foam.
Ethanol (150 mL) and 2 g of 10% palladium on activated carbon were added and the mixture was hydrogenated at 40 psi H2 overnight. TLC indicated formation of two new slower, closely-migrating compounds.
The catalyst was removed by filtration, and the filtrate concentrated in vacuo to an oily yellow foam. Silica gel flash chromatography (500 mL silica, eluted with 0-3% CH3 OH in CH2 Cl2) provided 1.85 g of semi-pure product, which was dissolved in 10 mL CH2 Cl2. A solid quickly precipitated, which was collected by filtration and washed with fresh CH2 Cl2. Vacuum desiccation yielded 1.5 g of product 13 (11% yield from 9 and 9i) as free white crystals. Mass-spectrometry, FAB+, M+H+, calculated: 453.1574, observed: 453.1574. 1 H NMR δ 8.21 (d, J=7.5 Hz, 1H), 8.08-8.13 (mm, 3H), 7.94 (d, J=7.4 Hz, 2H), 7.46-7.7 (mm, 8H), 6.04 (d, J=16.9 Hz, 1H), 5.08 (dd, J=3.6, 51.5 Hz, 1H), 4.85 (dd, J=3.3, 12.8 Hz, 1H), 4.80 (dr, J=2.1, 12.8 Hz, 1H), 4.26 (m, 1H), 3.48 (dm, J=27 Hz, 1H); 19 F N/VIR δ -200.1 (m).
N4,5 '-O-dibenzoyl-2'-fluoro-3'-(4-methoxytrityl) amino-2',3'-dideoxycytidine (compound 14, FIG. 3) was prepared as follows: To 0.9 g (2.0 mmol) of 13 in 25 mL anhydrous pyridine was added 0.86 g (2.8 mmol) 4-methoxytrityl chloride, and the mixture stirred overnight. The reaction was quenched with 0.5 mL H2 O and concentrated in vacuo. CH2 Cl2 (50 mL) was added and washed with 50 mL saturated aqueous NaHCO3 and with water (2×50 mL). The solvent was removed in vacuo, replaced with 10 mL CH2 Cl2, and pipetted into 80 mL rapidly stirred 1/1 hexane/ether. After further stirring for 2 h, the product was collected by filtration and dried under vacuum, giving 1.3 g (88% yield) of product as a white powder. Mass-spectrometry, FAB+, M+H+, calculated: 725.2775, observed: 725.2761. 1 H NMR δ 8.59 (br s, 1H), 8.07 (br d, J=5.7 Hz, 1H), 7.89 (br d, J=7 Hz, 2H), 7.83 (dd, J=1.3, 6.7 Hz, 2H), 7.68 (dd, J=7.4, 7.4 Hz, 2H), 7.5-7.6 (m, 8H), 7.43 (dd, J=2.1, 6.9 Hz, 2H), 7.1-7.3 (mm, 7H), 6.71 (d, J=8.9 Hz, 2H), 5.80 (d, J=15.4 Hz, 1H), 5.03 (dd,J=2.0, 13.0 Hz, 1H), 4.98 (dd, J=2.3;, 13.1 Hz, 1H), 4.41 (br d, J=10.5 Hz, 1H), 3.63 (s, 3H), 3.36 (dddd, J=3.1, 11.1, 11.1, 25.7 Hz, 1H), 2.84 (dd, J=3.1, 49.9 Hz, 1H), 2.52 (dd, J=2.7, 11.5 Hz, 1H); 19 F NMR δ -196.3 (m).
N4 -benzoyl-2'-fluoro-3'-(4-methoxytrityl) amino-2',3'-dideoxycytidine (compound 15, FIG. 3) was prepared as follows: To 1.3 g (1.75 mmol) of 14 in 20 mL of 65/30/5 pyridine/methanol/water, cooled in an ice bath, was added 10 mL of cold 2M NaOH in 65/30/5 pyrridine/methanol/water. The mixture was stirred cold for 20 min, then neutralized with pyridinium-H+ form Bio-Rad AG® 50W-X8 cation exchange resin. After 5 min, the resin was removed by filtration and washed with methanol. The combined filtrate and wash were concentrated in vacuo to an oil, which was dissolved in 100 mL ethyl acetate. The mixture was washed with 100 mL saturated aqueous NaHCO3 and with water (2×100mL). After concentration in vacuo to a foam, the product was dissolved in 10 mL CH2 Cl2 and pipetted into 75 mL rapidly stirred hexane/ether, 2/1. The product was collected by filtration and dried under vacuum, giving 1.13 g (102% yield) of product as a white powder. Mass-spectrometry, FAB+, M+Cs+, calculated: 753.1489, observed: 753.1499. 1 H NMR δ 8.30 (br d, J=6.8 Hz, 1H), 7.89 (br d, J=6.7 Hz, 2H), 7.64 (dd, J=7.4, 7.4 Hz, 1H), 7.44-7.56 (mm, 9H), 7.22-7.32 (mm, 9H), 6.82 (d, J=8.8 Hz, 2H), 5.80 (d, J=15.7 Hz, 1H), 4.26 (mm, 2H), 4.13 (d, J=10.2 Hz, 1H), 3.81 (s, 3H), 3.26 (dddd, J=3.4, 10.7, 10.8, 26.5 Hz, 1H), 2.93 (dd, J=3.3, 50.5 Hz, 1H), 2.50 (dd, J=2.8, 11.0 Hz, 1H); 19 F NMR δ -195.3 (m).
N4 -benzoyl-2'-fluoro-3'-(4-methoxytrityl)amino-2',3'-dideoxycytidine 5'-(2-cyanoethyl N,N-diisopropyl)phosphoramidite (compound 2c, FIG. 3) was prepared as follows: To 970 mg (1.56 mmol) of 15 in 25 mL anhydrous CH2 Cl2 was added 200 mg (1.17 mmol) of diisopropylammonium tetrazolide and 1.0 mL (3.15 mmol) of 2-cyanoethyl N,N,N',N'-tetraisopropylphosphorodiamidite. After stirring the mixture for 3 h, the solvent was removed in vacuo and the residue purified on a Chromatotron, using 4 mm plates and eluting with 0-1.5% methanol in 0.5% triethylamine in CH2 Cl2. The product was concentrated in vacuo to a foam, which was dissolved in 10 mL CH2 Cl2, and precipitated by slow addition to 40 mL of rapidly stirred hexane. After decanting the supernatant, the product was vacuum desiccated over P2 O5, giving 880 mg (69%) of white powder. Mass-spectrometry, FAB+, M+Cs+, calculated: 953.2568, observed: 953.2531. 19 F NMR δ -193.6 (m); 31 P NMR δ 150.4, 149.4.
EXAMPLE 4
N4 -benzoyl-2'-fluoro-3'-(4-methoxytrityl)amino-2',3'-dideoxycytidine 5'-succinyl-loaded CPG
Intermediate 15 was 5'-succinylated and loaded upon CPG solid support by standard procedures..sup. 15,16 More, particularly, N4 -benzoyl-2'-fluoro-3'-(4-methoxytrityl)amino-2',3'-dideoxycytidine 5'-succinyl-loaded CPG was prepared as follows: To 100 mg (0.16 mmol) of 15 in 2 mL anhydrous CH2 Cl2 was added 55 mg (0.55 mmol) of succinic anhydride and 65 mg (0.53 mmol) of dimethylaminopyridine. The mixture was stirred for 2 h, the evaporated in vacuo to an oil. The oil was dissolved in 20 mL CH2 Cl2, washed with 20 mL of saturated aqueous NaHCO3 and with water (2×20mL), and then reconcentrated in vacuo to a foam. To the foam was added 1 mL 0.4M diisopropylethylamine in DMSO/N-methylpyrrolidine, 1/1, and 0.7 mL 0.2M 1-hydroxybenzotriazole, 0.2M 2-(1H-benzotriazol-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate in DMSO/N-methylpyrrolidine, 1/1. After 3 min, the mixture was drawn into a 10 mL syringe containing 1.2 g of long-chain alkylamino-CPG. An additional 5 mL DMSO wash was also drawn into the syringe. The CPG-nucleoside mixture was mixed for 1.5 h, then the CPG washed with 5 volumes of anhydrous acetonitrile. Unreacted CPG amino groups were acetylated by standard ABI capping solutions for 2 min. The CPG was again washed with 5 volumes of acetonitrile and 5 volumes of CH2 Cl2. Nucleoside loading was determined to be approximately 5 μmole/g by standard trityl assay.
EXAMPLE 5
Solid Phase Synthesis of Oligo-2'-fluoronucleotide N3'→P5' Phosphoramidates
Oligo-2'-fluoronucleotide N3'→P5' phosphoramidates were synthesized on solid phase supports using either 5'-DMT-2'-fluoro-3'-aminonucleoside monomers (Scheme 1) or phosphoramidite monomers (of Schemes 2 and 3).
Dimer dUf np T was prepared using monomer of Scheme 1 via carbon tetrachloride-driven oxidative phosphorylation of dUf n by a 5'-H-phosphonate of the anchored dT. Product was analyzed and isolated by RP HPLC in 70% yield and the structure was confirmed by mass spectrometry and by acid catalyzed hydrolysis, which gave 2'-fluoro-3'-amino-2'-deoxyuridine and 5'-thymidylic acid. The same synthetic strategy was also used to introduce one or two 2'-fluoro-3'-aminonucleosides into longer oligonucleotide phosphoramidate chains. Compounds 20 and 21, Table 1, containing one or two 2'-fluoronucleosides in the middle of the chain were prepared and isolated by IE HPLC. Coupling yields of the 2'-fluoronucleoside 1 did not exceed 70-75% (in contrast to 94-96% for the 2'-deoxynucleosides), as judged by step-wise measurement of released DMT-cation and by IE HPLC analysis.
Compounds 22-25 (Table 1) were synthesized by way of phosphoramidite monomers. The average coupling efficiency as determined by released MMT-cation assay was ˜94% with single coupling per cycle and ˜96% with double application of Step 2 per synthetic cycle. A representative IE HPLC profile of a crude oligomer synthesis is shown in FIG. 6.
EXAMPLE 6
Thermal Stability of Duplexes of RNA or DNA and Oligo-2'-fluoronucleotide Phosphoramidates
The ability of the oligo-2'-fluoronucleoside phosphoramidates to hybridize with complementary DNA and RNA was evaluated. Melting temperatures were determined for duplexes formed under close to physiological salt concentrations and the results are summarized in Table 1. Substitution of one 2'-deoxynucleoside by one 2'-fluoronucleoside in a phosphoramidate decamer lead to an increase in Tm by ˜2° C. for either DNA or RNA hybrids (expt. 2 and 3, Table 1). Accordingly, replacement of two 2'-deoxynucleosides with 2'-fluoronucleosides in the same phosphoramidate decamer led to an increase in duplex's Tm of 3.5°-6.4° C. In contrast, others have reported that substitution with two central 2'-fluoronucleosides destabilizes the duplexes of phosphodiester oligomers.17
Substitution of all 2'-fluoronucleosides for 2'-deoxynucleosides in the decamer phosphoramidate resulted in significant enhancement of duplex thermal stability: Tm values were increased by 16°-25° C. (compare expt. 2 and 5, Table 1). The presented data show that further increases in the proportion of N-sugar ring pucker in the N3'→P5' phosphoramidates as well as additional negative polarization of 3'-amino groups (by 2'-fluorine) substantially stabilized oligonucleotide duplexes. It is noteworthy that Tm 's of the duplexes formed by 2'-fluoroamidates were 38°-44° C. higher than those of isosequential phosphodiester compounds, with 4°-5° C. per modification increase in melting temperatures (compare expt 1, 5, and 6, Table 1). Addition of magnesium cation to the melting buffer stabilized duplexes formed by phosphoramidates with DNA to a greater extent than those formed with RNA, suggesting a probable different secondary structure and solvation of the complexes being formed.
The same trend in duplex thermal stability was observed for mixed-base 11-mer 25 (Table 1), which formed more stable hybrids with complementary DNA and RNA than did the analogous oligo-2'-deoxynucleoside phosphoramidate 24 (compare expt. 7 and 8, Table 1).
                                  TABLE 1                                 
__________________________________________________________________________
Oligonucleotides and T.sub.m values of their duplexes.                    
                         T.sub.m, °C..sup.b                        
Expt Oligonucleotide.sup.a                                                
                         DNA.sup.c                                        
                                RNA.sup.c                                 
__________________________________________________________________________
1    UUUUUUUUUT, 18      16.7; 24.6                                       
                                17.9; 20.3                                
2    U.sub.np U.sub.np U.sub.np U.sub.np U.sub.np U.sub.np U.sub.np       
     U.sub.np U.sub.np T, 19                                              
                         18.5; 38.2                                       
                                38.1; 47.2                                
3    U.sub.np U.sub.np U.sub.np U.sub.np U.sup.f.sub.np U.sub.np U.sub.np 
     U.sub.np U.sub.np T, 20                                              
                         20.0; 41.0                                       
                                40,1; 49.3                                
4    U.sub.np U.sub.np U.sub.np U.sup.f.sub.np U.sup.f.sub.np U.sub.np    
     U.sub.np U.sub.np U.sub.np T, 21                                     
                         23.4; 44.6                                       
                                44.5; 52.7                                
5    .sub.p U.sup.f.sub.np U.sup.f.sub.np U.sup.f.sub.np U.sup.f.sub.np   
     U.sup.f.sub.np U.sup.f.sub.np U.sup.f.sub.np U.sup.f.sub.np U.sup.f.s
     ub.np T, 22         34.6; 63.0                                       
                                55.2; 64.6                                
6    .sub.p U.sup.f.sub.np U.sup.f.sub.np U.sup.f.sub.np U.sup.f.sub.np   
     U.sup.f.sub.np U.sup.f.sub.np U.sup.f.sub.np U.sup.f.sub.np U.sup.f.s
     ub.np U.sup.f.sub.n, 23                                              
                         39.5; 63.2                                       
                                56.4; 64.0                                
7    C.sub.np U.sub.np U.sub.np C.sub.np U.sub.np U.sub.np C.sub.np       
     C.sub.np U.sub.np U.sub.np A, 24                                     
                         44.2; --                                         
                                66.0; --                                  
8    C.sup.f.sub.np U.sup.f.sub.np U.sup.f.sub.np C.sup.f.sub.np U.sup.f.s
     ub.np U.sup.f.sub.np C.sup.f.sub.np C.sup.f.sub.np U.sup.f.sub.np    
     U.sup.f.sub.np A, 25                                                 
                         56.9; --                                         
                                81.6; --                                  
__________________________________________________________________________
 .sup.a All 2deoxy compounds; np, f, p, and n represent 3NHP(O)(O.sup.-)O5
 internucleoside link, 2fluorine, 5phosphate, and 3amine, respectively;   
 .sup.b T.sub.m was determined with 3 μM of oligonucleotides; first    
 values were determined in 10 mM sodium phosphate, 150 mM sodium chloride,
 pH 7.04; second values were determined in same buffer containing an      
 additional 10 mM magnesium chloride; dashes are for not determined T.sub.
 s.                                                                       
 .sup.c complementary target; poly(dA) or poly(rA) for experiments 1-6,   
 d(ATAAGGAAGAAGC) or r(AUAAGGAAGAAGC) for experiments 7, 8.               
EXAMPLE 7
Hydrolytic Stability of Oligo-2'-fluoronucleotide Phosphoramidates
Stability of the oligo-2'-fluoro- in comparison with the oligo-2'-deoxynucleotide N3'→P5' phosphoramidates toward enzymatic hydrolysis was evaluated. Phosphoramidates 19 and 22 (Table 1) were treated with snake venom phosphodiesterase and alkaline phosphatase, and analyzed at successive time points by IE HPLC. Under the conditions used, oligo-2'-deoxyphosphoramidate 19 and oligo-2'-fluorophosphoramidate 22 were hydrolyzed progressively and at similar rates, with calculated half-lives of the full-length strands equal to 4.9 and 5.4 h, respectively. In comparison, decathymidilic acid with natural phosphodiester linkages was completely digested to thymidine within 10 min under the same conditions.
The stability of the phosphoramidates toward acid-catalyzed hydrolysis was studied as follows. Oligonucleotides 19 and 22 were incubated at room temperature in 10% acetic acid, pH 2.2 or in 20 mM sodium acetate buffers, pH 4.7 and 5.3. The hydrolysis reactions were monitored by IE HPLC and the data are summarized in Table 2. The observed half-lives of full-length oligonucleotide 19 at pH 2.2, 4.7, and 5.3 were 21.5 min, 12.3 h, and 68 h, respectively. The oligo-2'-fluorophosphoramidate 22 was noticeably more stable under these conditions, with respective half-lives of this full-length oligomer of 61 min, 66 h, and 309 h. These results demonstrate a markedly reduced 3'-nitrogen basicity due to electron-withdrawing 2'-fluorine, with consequently greater acid stability of oligo-2'-fluorophosphoramidates relative to the parent oligo-2'-deoxyphosphoramidates.
              TABLE 2                                                     
______________________________________                                    
Acid Stability of the Oligonucleotide N3'→P5' Phosphoramidates     
                      T.sub.1/2, h                                        
                      pH                                                  
Expt Oligonucleotide        2.2    4.7  5.3                               
______________________________________                                    
1    U.sub.np U.sub.np U.sub.np U.sub.np U.sub.np U.sub.np U.sub.np       
     U.sub.np U.sub.np T, 19                                              
                            0.34   12.3 68                                
2    .sub.p U.sup.f.sub.np U.sup.f.sub.np U.sup.f.sub.np U.sup.f.sub.np   
     U.sup.f.sub.np U.sup.f.sub.np U.sup.f.sub.np U.sup.f.sub.np U.sup.f.s
     ub.np T, 22            1.0    66   309                               
______________________________________                                    
Cited References
1. (a) Helene, C.; Toulme, J. J. Biochem. Biophys. Acta 1990, 1049, 99-125.
(b) Uhlman, E.; Peyman, A. Chem. Rev. 1990, 90, 544-584
2. (a) Bielinska, A.; Shivdasami, R. A.; Zhang, L.; Nabel, G. J. Science 1990, 250, 997-101.
(b) Wyatt, J. R.; Vickers, T. A.; Roberson, J. L.; Buckheit, R. W. Jr.; Klimkait, T.; DeBeats, E.; Davis, P. W.; Rayner, B.; Imbach, J. L.; Ecker, D. J. Proc. Natl. Acad. Sci., USA 1994, 91, 1356-1360.
3. DeMesmacker, A.; Altman, K. H.; Waldner, A.; Wendebom, S. Curr Opinion Struct. Biol. 1995, 5, 343-355.
4. (a) Gryaznov, S.M.; Chen, I. K. J. Am. Chem. Soc. 1994, 116, 3143-3144.
(b) Gryaznov, S. M.; Lloyd, D. H.; Chen, J. K.; Schultz, R. G.; DeDionisio, L. A.; Ratmeyer, L.; Wilson, W. D. Proc. Natl. Acad. Sci., USA 1995, 92, 5798-02.
(c) Chen, J. K.; Schultz, R. G.; Lloyd, D. H.; Gryaznov, S. M. Nucleic Acids Res. 95, 23, 2661-2668.
5. Ding, D.; Gryaznov, S. M.; Lloyd, D. H.; Chandrasekaran, S.; Yao, S.; Ratmeyer, L.; Li Pan, Y.; Wilson, W. D. Nucleic Acids Res. 1996, 24, in press.
6. Gryaznov, S. M.; Letsinger, R. L. Nucleic Acids Res. 1992, 20, 3403-3409.
7. (a) Kawasaki, A. M.; Casper, M. D.; Freier, S. M. Lesnik, E. A.; Zounes, M. C.; Cummins, L. L.; Gonzalez, C.; Cook, P. D. J. Med. Chem. 1993, 36, 831-841.
(b) Monia, B. P.; Lesnik, E. A.; Gonzalez, C.; Lima, W. F.; McGee, D.; Guinosso, C. J.; Kawasaki, A. M.; Cook, P. D.; Freier, S .M. J. Biol. Chem. 1993, 268, 514-14522.
8. Pieken, W. A.; Olsen, D. B.; Benseler, F.; Aurup, K.; H.; Eckstein, F. Science 91, 253, 314-317.
9. (a) Ikehara, M. Heterocycles 1984, 21, 75-90.
(b) Guschlbauer, W.; Jankowski, K. Nucleic Acids Res. 1980, 8, 1421-1433.
10. Webb, T. R.; Mitsuya, H.; Broder, S. J. Med. Chem. 1988, 31, 1475-1479.
11. Codington, J. F.; Fecher, R.; Fox, J. J. J. Am. Chem. Soc. 1960, 82, 2794-03.
12. Codington, J. F.; Fecher, R.; Fox, J. J. J. Organic Chem. 1962, 27, 163-167.
13. Reichman, U.; Hollenberg, D. H.; Chu, C. K.; Watanabe, K. A.; Fox, J. J. J. Organic Chem. 1976, 41, 2042-2043.
14. Divakar, K. J.; Reese, C. B. J. Chem. Soc., Perkin. Trans. 1 1982, 1171-76.
15. Atkinson, T.; Smith, M. In Oligonucleotide Synthesis. A Practical Approach, Gait, M. J. Ed., IRL Press, 1984, 35-81.
16. Knorr, R.; Trzeciak, A.; Bannwarth, W.; Gillessen, D. Tetrahedron Lat. 1989, 30, 1927-1930.
17. Williams, D. M.; Beuseler, F.; Eckstein, F. Biochemistry 1991, 30, 4001-09.
18. Caruthers, M. H. Accounts Chem. Res. 1991, 24, 278-284.
19. Gryaznov, S. M.; Letsinger, R. L. Nucleic Acids Res. 1993, 21, 1403-1408.
20. Obenland, C. O.; Mangold, D. J.; Marino, M. P. Inorg. Synth. 1966, 8, 53-56.
__________________________________________________________________________
SEQUENCE LISTING                                                          
(1) GENERAL INFORMATION:                                                  
(iii) NUMBER OF SEQUENCES: 4                                              
(2) INFORMATION FOR SEQ ID NO: 1:                                         
(i) SEQUENCE CHARACTERISTICS:                                             
(A) LENGTH: 10 nucleotides                                                
(B) TYPE: nucleic acid                                                    
(C) STRANDEDNESS: single                                                  
(D) TOPOLOGY: linear                                                      
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 1:                                  
UUUUUUUUUT10                                                              
(2) INFORMATION FOR SEQ ID NO: 2:                                         
(i) SEQUENCE CHARACTERISTICS:                                             
(A) LENGTH: 11 nucleotides                                                
(B) TYPE: nucleic acid                                                    
(C) STRANDEDNESS: single                                                  
(D) TOPOLOGY: linear                                                      
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 2:                                  
CUUCUUCCUUA11                                                             
(2) INFORMATION FOR SEQ ID NO: 3:                                         
(i) SEQUENCE CHARACTERISTICS:                                             
(A) LENGTH: 13 nucleotides                                                
(B) TYPE: nucleic acid                                                    
(C) STRANDEDNESS: single                                                  
(D) TOPOLOGY: linear                                                      
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 3:                                  
ATAAGGAAGAAGC13                                                           
(2) INFORMATION FOR SEQ ID NO: 4:                                         
(i) SEQUENCE CHARACTERISTICS:                                             
(A) LENGTH: 13 nucleotides                                                
(B) TYPE: nucleic acid                                                    
(C) STRANDEDNESS: single                                                  
(D) TOPOLOGY: linear                                                      
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 4:                                  
AUAAGGAAGAAGC13                                                           
__________________________________________________________________________

Claims (16)

We claim:
1. A polynucleotide comprising an oligo-2'-fluoronucleoside N3'→P5' phosphoramidate segment defined by the formula: ##STR4## wherein B is a purine or pyrimidine, n is an integer greater than 1, and R4 is hydrogen or a phosphate protecting group.
2. The polynucleotide of claim 1, wherein R4 is hydrogen or a phosphate protecting group selected from the group consisting of methyl, β-cyanoethyl, or 4-nitrophenylethyl.
3. The polynucleotide of claim 1, wherein n is an integer between 2 and 30.
4. The polynucleotide of claim 3, wherein n is an integer between 2 and 20.
5. The polynucleotide of claim 1, wherein B is a pyrimidine.
6. The polynucleotide of claim 1 in a triplex comprising said polynucleotide and a target double-stranded DNA.
7. The polynucleotide of claim 1, wherein the oligo-2'-fluoronucleoside N3'→P5' phosphoramidate segment is defined by the formula: ##STR5## wherein R1 is hydroxyl or a 5'-hydroxyl protecting group.
8. The polynucleotide of claim 7, wherein R1 is hydroxyl or a trityl group.
9. The polynucleotide of claim 7, wherein R4 is hydrogen or a phosphate protecting group selected from the group consisting of methyl, β-cyanoethyl, or 4-nitrophenylethyl.
10. The polynucleotide of claim 7, wherein n is an integer between 2 and 30.
11. The polynucleotide of claim 10, wherein n is an integer between 2 and 20.
12. The polynucleotide of claim 7, wherein B is a pyrimidine.
13. The polynucleotide of claim 7 in a triplex comprising said polynucleotide and a target double-stranded DNA.
14. The polynucleotide of claim 7, wherein R1 is hydroxyl, R4 is H, and n is an integer greater than 8.
15. The polynucleotide of claim 14, wherein n is an integer between 8 and 20.
16. The polynucleotide of claim 15, hybridized to double-stranded DNA to form a triplex.
US08/603,566 1996-02-21 1996-02-21 Oligo-2'-fluoronucleotide N3'->P5' phosphoramidates Expired - Lifetime US5684143A (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
US08/603,566 US5684143A (en) 1996-02-21 1996-02-21 Oligo-2'-fluoronucleotide N3'->P5' phosphoramidates
DE69619141T DE69619141T2 (en) 1996-02-21 1996-06-14 SOLID PHASE SYNTHESIS OF OLIGONUCLEOTIDE N3'-P5'-PHOSPHORAMIDATES
PCT/US1996/010418 WO1997031009A1 (en) 1996-02-21 1996-06-14 Solid phase synthesis of oligonucleotide n3'→p5' phosphoramidates
US08/663,918 US5824793A (en) 1996-02-21 1996-06-14 Solid phase synthesis of oligonucleotide N3'-P5' phosphoramidates
CA 2245666 CA2245666C (en) 1996-02-21 1996-06-14 Solid phase synthesis of oligonucleotide n3'.fwdarw.p5' phosphoramidates
PL96328639A PL183661B1 (en) 1996-02-21 1996-06-14 Synthesis of p5' phosphoamidane oligonucleotides n3'
CZ982629A CZ262998A3 (en) 1996-02-21 1996-06-14 Synthesis process of oligonucleotide n3 - p5 phosphoramidates on solid phase
AT96919449T ATE212997T1 (en) 1996-02-21 1996-06-14 SOLID PHASE SYNTHESIS OF OLIGONUCLEOTIDE N3'-P5' PHOSPHORAMIDATE
AU61789/96A AU703509C (en) 1996-02-21 1996-06-14 Solid phase synthesis of oligonucleotide N3'-P5' phosphoramidates
EP96919449A EP0882059B1 (en) 1996-02-21 1996-06-14 Solid phase synthesis of oligonucleotide n3-'p5'- phosphoramidates
US08/771,789 US5859233A (en) 1996-02-21 1996-12-20 Synthons for synthesis of oligonucleotide N3-P5 phosphoramidates

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US08/603,566 US5684143A (en) 1996-02-21 1996-02-21 Oligo-2'-fluoronucleotide N3'->P5' phosphoramidates

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US08/663,918 Continuation-In-Part US5824793A (en) 1996-02-21 1996-06-14 Solid phase synthesis of oligonucleotide N3'-P5' phosphoramidates

Publications (1)

Publication Number Publication Date
US5684143A true US5684143A (en) 1997-11-04

Family

ID=24415986

Family Applications (2)

Application Number Title Priority Date Filing Date
US08/603,566 Expired - Lifetime US5684143A (en) 1996-02-21 1996-02-21 Oligo-2'-fluoronucleotide N3'->P5' phosphoramidates
US08/663,918 Expired - Lifetime US5824793A (en) 1996-02-21 1996-06-14 Solid phase synthesis of oligonucleotide N3'-P5' phosphoramidates

Family Applications After (1)

Application Number Title Priority Date Filing Date
US08/663,918 Expired - Lifetime US5824793A (en) 1996-02-21 1996-06-14 Solid phase synthesis of oligonucleotide N3'-P5' phosphoramidates

Country Status (7)

Country Link
US (2) US5684143A (en)
EP (1) EP0882059B1 (en)
AT (1) ATE212997T1 (en)
CZ (1) CZ262998A3 (en)
DE (1) DE69619141T2 (en)
PL (1) PL183661B1 (en)
WO (1) WO1997031009A1 (en)

Cited By (79)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001053307A1 (en) * 2000-01-21 2001-07-26 Geron Corporation 2'-arabino-fluorooligonucleotide n3'→p5'phosphoramidates: their synthesis and use
US6342214B1 (en) * 1995-05-16 2002-01-29 Karl Tryggvason Method for viral vector delivery
US6440723B1 (en) 1998-12-30 2002-08-27 Oligos Etc. Inc. Arrays with modified oligonucleotide and polynucleotide compositions
US20020137074A1 (en) * 2000-11-21 2002-09-26 Piunno Paul A.E. Selectivity of nucleic acid diagnostic and microarray technologies by control of interfacial nucleic acid film chemistry
US6503711B1 (en) * 1997-06-18 2003-01-07 Ulrich J. Krull Nucleic acid biosensor diagnostics
US6608036B1 (en) 1999-09-10 2003-08-19 Geron Corporation Oligonucleotide N3′→P5′ thiophosphoramidates: their synthesis and administration to treat neoplasms
US6638264B1 (en) 1995-05-16 2003-10-28 Biostratum Incorporation Perfusion apparatus and methods for pharmaceutical delivery
WO2004060323A2 (en) * 2003-01-02 2004-07-22 The Research Foundation Of State University Of Newyork Novel inhibitors of poxvirus replication
WO2005054494A2 (en) 2003-11-26 2005-06-16 University Of Massachusetts Sequence-specific inhibition of small rna function
WO2005089287A2 (en) 2004-03-15 2005-09-29 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
US20070270370A1 (en) * 1999-07-22 2007-11-22 Sankyo Company, Limited Novel bicyclonucleoside analogues
US20090022667A1 (en) * 2007-05-15 2009-01-22 Marco Peters METHODS OF TREATING COGNITIVE DISORDERS BY INHIBITION OF Gpr12
US20090105468A1 (en) * 2004-07-02 2009-04-23 Gryaznov Sergei M Synthesis of Protected 3'-Amino Nucleoside Monomers
US20100029008A1 (en) * 2004-12-16 2010-02-04 Febit Biotech Gmbh Polymerase-independent analysis of the sequence of polynucleotides
US7695902B2 (en) 1996-06-06 2010-04-13 Isis Pharmaceuticals, Inc. Oligoribonucleotides and ribonucleases for cleaving RNA
US20100173863A1 (en) * 1998-02-25 2010-07-08 Schinazi Raymond F 2-Fluoronucleosides
WO2010100404A2 (en) 2009-03-02 2010-09-10 Mina Therapeutics Limited Rna molecules and therapeutic uses thereof
WO2010105096A2 (en) 2009-03-11 2010-09-16 University Of Massachusetts Modulation of human cytomegalovirus replication by micro-rna 132 (mir132), micro-rna 145 (mir145) and micro-rna 212 (mir212)
US20100240441A1 (en) * 2007-09-14 2010-09-23 Konami Digital Entertainment Co., Ltd Game system, and game apparatus and challenge notifying apparatus constituting the game system
US7812149B2 (en) 1996-06-06 2010-10-12 Isis Pharmaceuticals, Inc. 2′-Fluoro substituted oligomeric compounds and compositions for use in gene modulations
US7884086B2 (en) 2004-09-08 2011-02-08 Isis Pharmaceuticals, Inc. Conjugates for use in hepatocyte free uptake assays
EP2314692A2 (en) 2003-06-02 2011-04-27 University of Massachusetts Methods and compositions for enhancing the efficacy and specificity of RNAi
WO2011075188A1 (en) 2009-12-18 2011-06-23 Dicerna Pharmaceuticals, Inc. Dicer substrate agents and methods for the specific inhibition of gene expression
WO2012129363A2 (en) 2011-03-24 2012-09-27 President And Fellows Of Harvard College Single cell nucleic acid detection and analysis
US8394947B2 (en) 2004-06-03 2013-03-12 Isis Pharmaceuticals, Inc. Positionally modified siRNA constructs
WO2013075132A1 (en) 2011-11-17 2013-05-23 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Therapeutic rna switches compositions and methods of use
US8569474B2 (en) 2004-03-09 2013-10-29 Isis Pharmaceuticals, Inc. Double stranded constructs comprising one or more short strands hybridized to a longer strand
US8604183B2 (en) 2002-11-05 2013-12-10 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2′-modified nucleosides for use in gene modulation
US8664189B2 (en) 2008-09-22 2014-03-04 Rxi Pharmaceuticals Corporation RNA interference in skin indications
WO2014113540A1 (en) 2013-01-16 2014-07-24 Iowa State University Research Foundation, Inc. A deep intronic target for splicing correction on spinal muscular atrophy gene
US8815818B2 (en) 2008-07-18 2014-08-26 Rxi Pharmaceuticals Corporation Phagocytic cell delivery of RNAI
EP2821085A2 (en) 2003-09-12 2015-01-07 University of Massachusetts RNA interference for the treatment of gain-of-function disorders
US9074211B2 (en) 2008-11-19 2015-07-07 Rxi Pharmaceuticals Corporation Inhibition of MAP4K4 through RNAI
US9080171B2 (en) 2010-03-24 2015-07-14 RXi Parmaceuticals Corporation Reduced size self-delivering RNAi compounds
US9096636B2 (en) 1996-06-06 2015-08-04 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
US9133233B2 (en) 2003-11-04 2015-09-15 Geron Corporation RNA amidates and thioamidates for RNAi
US9228189B2 (en) 2012-10-26 2016-01-05 Geron Corporation C-myc antisense oligonucleotides and methods for using the same to treat cell-proliferative disorders
US9340786B2 (en) 2010-03-24 2016-05-17 Rxi Pharmaceuticals Corporation RNA interference in dermal and fibrotic indications
WO2016100401A1 (en) 2014-12-15 2016-06-23 Dicerna Pharmaceuticals, Inc. Ligand-modified double-stranded nucleic acids
US9388415B2 (en) 2003-09-09 2016-07-12 Geron Corporation Modified oligonucleotides for telomerase inhibition
WO2016161378A1 (en) 2015-04-03 2016-10-06 University Of Massachusetts Oligonucleotide compounds for treatment of preeclampsia and other angiogenic disorders
WO2016161374A1 (en) 2015-04-03 2016-10-06 University Of Massachusetts Oligonucleotide compounds for targeting huntingtin mrna
WO2016161388A1 (en) 2015-04-03 2016-10-06 University Of Massachusetts Fully stabilized asymmetric sirna
WO2016172346A1 (en) 2015-04-23 2016-10-27 Geron Corporation Methods of polynucleotide preparation using multivalent cation salt compositions
US9493774B2 (en) 2009-01-05 2016-11-15 Rxi Pharmaceuticals Corporation Inhibition of PCSK9 through RNAi
WO2017015075A1 (en) 2015-07-17 2017-01-26 President And Fellows Of Harvard College Methods of amplifying nucleic acid sequences
WO2017049192A1 (en) 2015-09-17 2017-03-23 University Of Massachusetts Compositions and methods for modulating fmr1 expression
US9745574B2 (en) 2009-02-04 2017-08-29 Rxi Pharmaceuticals Corporation RNA duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
US9796747B2 (en) 2014-05-01 2017-10-24 Geron Corporation Oligonucleotide compositions and methods of making the same
US10131904B2 (en) 2008-02-11 2018-11-20 Rxi Pharmaceuticals Corporation Modified RNAi polynucleotides and uses thereof
US10184124B2 (en) 2010-03-24 2019-01-22 Phio Pharmaceuticals Corp. RNA interference in ocular indications
EP3444350A1 (en) 2013-07-03 2019-02-20 Dicerna Pharmaceuticals, Inc. Methods and compositions for the specific inhibition of alpha-1 antitrypsin by double-stranded rna
EP3502252A1 (en) 2003-06-02 2019-06-26 University of Massachusetts Methods and compositions for controlling efficacy of rna silencing
US10443055B2 (en) 2016-12-22 2019-10-15 Beth Israel Deaconess Medical Center Compounds that target MYC microRNA responsive elements for the treatment of MYC-associated cancer
US10478503B2 (en) 2016-01-31 2019-11-19 University Of Massachusetts Branched oligonucleotides
US10556020B2 (en) 2014-09-26 2020-02-11 University Of Massachusetts RNA-modulating agents
US10633653B2 (en) 2015-08-14 2020-04-28 University Of Massachusetts Bioactive conjugates for oligonucleotide delivery
US10689647B2 (en) 2015-05-08 2020-06-23 Dicerna Pharmaceuticals, Inc. Methods and compositions for the specific inhibition of antithrombin 3 (AT3) by double-stranded RNA
EP3708680A1 (en) 2015-04-15 2020-09-16 The University of Massachusetts Compositions and methods for xi chromosome reactivation
US10808247B2 (en) 2015-07-06 2020-10-20 Phio Pharmaceuticals Corp. Methods for treating neurological disorders using a synergistic small molecule and nucleic acids therapeutic approach
US10900039B2 (en) 2014-09-05 2021-01-26 Phio Pharmaceuticals Corp. Methods for treating aging and skin disorders using nucleic acids targeting Tyr or MMP1
US10934550B2 (en) 2013-12-02 2021-03-02 Phio Pharmaceuticals Corp. Immunotherapy of cancer
US11001845B2 (en) 2015-07-06 2021-05-11 Phio Pharmaceuticals Corp. Nucleic acid molecules targeting superoxide dismutase 1 (SOD1)
US11021707B2 (en) 2015-10-19 2021-06-01 Phio Pharmaceuticals Corp. Reduced size self-delivering nucleic acid compounds targeting long non-coding RNA
EP3896451A1 (en) 2016-11-07 2021-10-20 University Of Massachusetts Carm1 inhibitors for facioscapulohumeral muscular dystrophy
US11279934B2 (en) 2014-04-28 2022-03-22 Phio Pharmaceuticals Corp. Methods for treating cancer using nucleic acids targeting MDM2 or MYCN
US11306312B2 (en) 2017-09-01 2022-04-19 Thomas Jefferson University Compositions and methods for MYC messenger RNA inhibitors
US11401543B2 (en) * 2012-09-27 2022-08-02 Bioo Scientific Corporation Methods and compositions for improving removal of ribosomal RNA from biological samples
US11401519B2 (en) 2017-06-07 2022-08-02 University Of Massachusetts Anti-ADAM33 oligonucleotides and related methods
US11629347B2 (en) 2019-05-06 2023-04-18 University Of Massachusetts Anti-C9ORF72 oligonucleotides and related methods
WO2023086292A2 (en) 2021-11-10 2023-05-19 University Of Rochester Gata4-targeted therapeutics for treatment of cardiac hypertrophy
WO2023086295A2 (en) 2021-11-10 2023-05-19 University Of Rochester Antisense oligonucleotides for modifying protein expression
US11702659B2 (en) 2021-06-23 2023-07-18 University Of Massachusetts Optimized anti-FLT1 oligonucleotide compounds for treatment of preeclampsia and other angiogenic disorders
WO2023164636A1 (en) 2022-02-25 2023-08-31 Vor Biopharma Inc. Compositions and methods for homology-directed repair gene modification
US11753638B2 (en) 2016-08-12 2023-09-12 University Of Massachusetts Conjugated oligonucleotides
WO2023196816A1 (en) 2022-04-04 2023-10-12 Vor Biopharma Inc. Compositions and methods for mediating epitope engineering
US11827882B2 (en) 2018-08-10 2023-11-28 University Of Massachusetts Modified oligonucleotides targeting SNPs
US11873487B2 (en) 2019-06-20 2024-01-16 University Of Massachusetts Compositions and methods for improved gene editing
WO2024050261A1 (en) 2022-08-29 2024-03-07 University Of Rochester Antisense oligonucleotide-based anti-fibrotic therapeutics

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6043070A (en) * 1997-08-29 2000-03-28 The Perkin-Elmer Corporation Phosphoramidate-phosphodiester oligonucleotide chimera as primers
US6958215B1 (en) 1999-03-15 2005-10-25 Geron Corporation Methods and compositions for inhibition of RNA splicing
AU5460700A (en) * 1999-06-04 2000-12-28 Allied-Signal Inc. Protected nucleosides
US6451998B1 (en) 1999-10-18 2002-09-17 Agilent Technologies, Inc. Capping and de-capping during oligonucleotide synthesis
US6881538B1 (en) 2000-03-05 2005-04-19 3M Innovative Properties Company Array comprising diamond-like glass film
US7563618B2 (en) 2001-03-23 2009-07-21 Geron Corporation Oligonucleotide conjugates
WO2004096286A2 (en) 2003-04-25 2004-11-11 Gilead Sciences, Inc. Antiviral phosphonate analogs
US7339052B2 (en) * 2005-02-08 2008-03-04 Honeywell International Inc. Phosphoramidite activator for oligonucleotide synthesis
CN103382212B (en) * 2013-07-06 2016-08-31 中国科学院成都生物研究所 5 ' position modified purine mycin compounds and its production and use
US9708360B2 (en) 2013-09-30 2017-07-18 Geron Corporation Phosphorodiamidate backbone linkage for oligonucleotides
US10072261B1 (en) * 2016-03-25 2018-09-11 Agilent Technologies, Inc. Double coupling method for oligonucleotide synthesis

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0490281A1 (en) * 1990-12-11 1992-06-17 Hoechst Aktiengesellschaft 3'- Amino or thiolmodified, fluorescence coupled nucleoside and oligonucleotide, a method for their preparation and their use
DE4129318A1 (en) * 1991-09-03 1993-03-04 Bayer Ag NEW ANTISENSE OLIGONUCLETIDES WITH CONTINUOUS PHOSPHORAMIDATE INTERNAL CLOTH BINDING
EP0552766A2 (en) * 1992-01-22 1993-07-28 Hoechst Aktiengesellschaft Oligonucleotide analogues, their preparation and use
US5476925A (en) * 1993-02-01 1995-12-19 Northwestern University Oligodeoxyribonucleotides including 3'-aminonucleoside-phosphoramidate linkages and terminal 3'-amino groups

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0490281A1 (en) * 1990-12-11 1992-06-17 Hoechst Aktiengesellschaft 3'- Amino or thiolmodified, fluorescence coupled nucleoside and oligonucleotide, a method for their preparation and their use
DE4129318A1 (en) * 1991-09-03 1993-03-04 Bayer Ag NEW ANTISENSE OLIGONUCLETIDES WITH CONTINUOUS PHOSPHORAMIDATE INTERNAL CLOTH BINDING
CA2077314A1 (en) * 1991-09-03 1993-03-04 Antonius Lobberding Antisense oligonucleotides with phosphoramidate internucleotide linkage throughout
EP0552766A2 (en) * 1992-01-22 1993-07-28 Hoechst Aktiengesellschaft Oligonucleotide analogues, their preparation and use
US5476925A (en) * 1993-02-01 1995-12-19 Northwestern University Oligodeoxyribonucleotides including 3'-aminonucleoside-phosphoramidate linkages and terminal 3'-amino groups

Non-Patent Citations (54)

* Cited by examiner, † Cited by third party
Title
Abraham, et al., "A Phosphoramidite-Based Synthesis of Phosphoramidate Amino Acid Diesters of Antiviral Nucleosides," Nucleosides & Nucleotides 13:1891-1903 (1994).
Abraham, et al., A Phosphoramidite Based Synthesis of Phosphoramidate Amino Acid Diesters of Antiviral Nucleosides, Nucleosides & Nucleotides 13:1891 1903 (1994). *
Agrawal, et al., "Oligodeoxynucleoside Phosphoramidates and Phosphoro-thioates as Inhibitors of Human Immunodeficiency Virus," Proc. Natl. Acad. Sci. USA 85:7079-7083 (1988).
Agrawal, et al., Oligodeoxynucleoside Phosphoramidates and Phosphoro thioates as Inhibitors of Human Immunodeficiency Virus, Proc. Natl. Acad. Sci. USA 85:7079 7083 (1988). *
Azhayev, et al., "Aminonucleosides and Their Derivatives. IX. Synthesis of Short Oligoribonucleotide with Phosphoramide Internucleotide Bonds," Bioorganicheskaya Khimiya 8:1218-1224 (1982). translation attached!.
Azhayev, et al., "Synthesis of Phosphoramidate Analogs of Ribonucleo-side Phosphates," Coll. Czechoslov. Chem. Commun. 44:792-798 (1979).
Azhayev, et al., "Synthesis of Phosphoramidate Analogues of Short Oligoribunucleotides," Nuc. Acids Res., Synm. Series No. 9, pp. 251-254 (1981).
Azhayev, et al., Aminonucleosides and Their Derivatives. IX. Synthesis of Short Oligoribonucleotide with Phosphoramide Internucleotide Bonds, Bioorganicheskaya Khimiya 8:1218 1224 (1982). translation attached . *
Azhayev, et al., Synthesis of Phosphoramidate Analogs of Ribonucleo side Phosphates, Coll. Czechoslov. Chem. Commun . 44:792 798 (1979). *
Azhayev, et al., Synthesis of Phosphoramidate Analogues of Short Oligoribunucleotides, Nuc. Acids Res., Synm. Series No. 9, pp. 251 254 (1981). *
Bannwarth, "166. Solid-Phase Synthesis of Oligodeoxynucleotides Containing Phosphoramidate Internucleotide Linkages and Their Specific Chemical Cleavage," Helvetica Chim. Acta 71:1517-1527 (1988).
Bannwarth, 166. Solid Phase Synthesis of Oligodeoxynucleotides Containing Phosphoramidate Internucleotide Linkages and Their Specific Chemical Cleavage, Helvetica Chim. Acta 71:1517 1527 (1988). *
Chen, et al., "Synthesis of Oligonucleotide N3'→P5' Phosphoramidates," Nuc. Acids Res. 23:2661-2668 (1995).
Chen, et al., Synthesis of Oligonucleotide N3 P5 Phosphoramidates, Nuc. Acids Res . 23:2661 2668 (1995). *
Dagle, et al., "Physical Properties of Oligonucleotides Containing Phosphoramide-Modified Internucleoside Linkages," Nuc. Acids Res. 19:1805-1810 (1991).
Dagle, et al., Physical Properties of Oligonucleotides Containing Phosphoramide Modified Internucleoside Linkages, Nuc. Acids Res . 19:1805 1810 (1991). *
Gryaznov, et al., "Oligonucleotide N3'→N5' Phosphoramidates," Proc. Natl. Acad. Sci. USA 92:5798-5802 (1995).
Gryaznov, et al., "Oligonucleotide N3'→N5' Phosphoramidates: Synthesis and Hybridization Properties," J. Am. Chem. Soc. 116:3143-3144 (1994).
Gryaznov, et al., "Synthesis and Properties of Oligonucleotides Containing Aminodeoxythymidine Units," Nuc. Acids Res. 20:3403-3409 (1992).
Gryaznov, et al., Oligonucleotide N3 N5 Phosphoramidates, Proc. Natl. Acad. Sci. USA 92:5798 5802 (1995). *
Gryaznov, et al., Oligonucleotide N3 N5 Phosphoramidates: Synthesis and Hybridization Properties, J. Am. Chem. Soc . 116:3143 3144 (1994). *
Gryaznov, et al., Synthesis and Properties of Oligonucleotides Containing Aminodeoxythymidine Units, Nuc. Acids Res . 20:3403 3409 (1992). *
Herdewijn, et al., "Synthesis and Biological Activity of the Mono- and Diamino Analogues of 2'-Deoxyadenosine, Cordycepin, 9-(3-deoxyβ-D-threopentofuranosyl)-adenine," Nucleosides & Nucleotides 8:1231-1257 (1989).
Herdewijn, et al., Synthesis and Biological Activity of the Mono and Diamino Analogues of 2 Deoxyadenosine, Cordycepin, 9 (3 deoxy D threopentofuranosyl) adenine, Nucleosides & Nucleotides 8:1231 1257 (1989). *
Imazawa, et al., "Synthesis of 3'-azido-2',3'-dideoxyribofuranosyl-purines," J. Org. Chem. 43:3045-3047 (1978).
Imazawa, et al., Synthesis of 3 azido 2 ,3 dideoxyribofuranosyl purines, J. Org. Chem . 43:3045 3047 (1978). *
Kawasaki, A. et al "Uniformly Modified 2'-Deoxy-fluoro Phosphorothioate Oligonucleotides as Nuclease-Resistant Antisense Compounds with High Affinity and Specifcity for RNA Targets." J. Med. Chem 1993 vol. 36 pp. 831-841.
Kawasaki, A. et al Uniformly Modified 2 Deoxy fluoro Phosphorothioate Oligonucleotides as Nuclease Resistant Antisense Compounds with High Affinity and Specifcity for RNA Targets. J. Med. Chem 1993 vol. 36 pp. 831 841. *
Krayevsky, et al., "Synthesis of Oligonucleotides with 5'→3' Phospho-amidoester Bond," Nuc. Acids Res., Sym. Series No. 9, pp. 203-205 (1981).
Krayevsky, et al., Synthesis of Oligonucleotides with 5 3 Phospho amidoester Bond, Nuc. Acids Res., Sym. Series No. 9, pp. 203 205 (1981). *
Mag, et al., "Synthesis and Selective Cleavage of an Oligodeoxynucleotide Containing a Bridged Non-Chiral Internucleotide 3'-Phosphoramidate Linkage," Tetrahedron Lett. 33:7319-7322 (1992).
Mag, et al., "Synthesis and Selective Cleavage of Oligodeoxyribonucleosides Containing Non-Chiral Internucleotide Phosphoramide Linkages," Nuc. Acids Res. 17:5973-5988 (1989).
Mag, et al., "Synthesis of Dinucleotides Containing a Bridged Non-Chiral Internucleoside 5'- or 3'-phosphoramidate Linkage," Tetrahedron 50: 10225-10234 (1994).
Mag, et al., Synthesis and Selective Cleavage of an Oligodeoxynucleotide Containing a Bridged Non Chiral Internucleotide 3 Phosphoramidate Linkage, Tetrahedron Lett . 33:7319 7322 (1992). *
Mag, et al., Synthesis and Selective Cleavage of Oligodeoxyribonucleosides Containing Non Chiral Internucleotide Phosphoramide Linkages, Nuc. Acids Res. 17:5973 5988 (1989). *
Mag, et al., Synthesis of Dinucleotides Containing a Bridged Non Chiral Internucleoside 5 or 3 phosphoramidate Linkage, Tetrahedron 50: 10225 10234 (1994). *
Milligan, et al., "Current Concepts in Antisense Drug Design," J. Med. Chem. 36:1923-1937 (1993).
Milligan, et al., Current Concepts in Antisense Drug Design, J. Med. Chem. 36:1923 1937 (1993). *
Pieken, W. et al "Kinetic Characterization of Ribonuclease Resistant 2'-Modified Hammerhead Ribozymes." Science vol. 253 pp. 314-317, 1991.
Pieken, W. et al Kinetic Characterization of Ribonuclease Resistant 2 Modified Hammerhead Ribozymes. Science vol. 253 pp. 314 317, 1991. *
Saha, et al., "Synthesis of Modified Achiral Internucleoside Linkages: NCHCH2 CH2 -Linked Oligonucleosides," Tetrahedron Lett. 34:6017-6020 (1993).
Saha, et al., Synthesis of Modified Achiral Internucleoside Linkages: NCHCH 2 CH 2 Linked Oligonucleosides, Tetrahedron Lett . 34:6017 6020 (1993). *
Ulhmann, et al., "Antisense Oligonucleotides: a New Therapeutic Principal," Chem. Rev. 90:543-584 (1990).
Ulhmann, et al., Antisense Oligonucleotides: a New Therapeutic Principal, Chem. Rev . 90:543 584 (1990). *
Williams, et al., "Properties of 2'-fluorothymidine-Containing Oligonucleotides: Interaction with Restriction Endonuclease EcoRV," Biochem. 30:4001-4009 (1991).
Williams, et al., Properties of 2 fluorothymidine Containing Oligonucleotides: Interaction with Restriction Endonuclease EcoRV , Biochem. 30:4001 4009 (1991). *
Zaitseva, et al., "Aminonucleosides and Their Derivatives. X. 2'-deoxydinucleoside Phosphates and Deoxydinucleotides with Phosphoamide Bonds," Bioorganicheskaya Khimiya 10:401-407 (1984). translation attached!.
Zaitseva, et al., Aminonucleosides and Their Derivatives. X. 2 deoxydinucleoside Phosphates and Deoxydinucleotides with Phosphoamide Bonds, Bioorganicheskaya Khimiya 10:401 407 (1984). translation attached . *
Zielinski, et al., "Autocatalytic Synthesis of a Tetranucleotide Analog," Nature 327:346-347 (1987).
Zielinski, et al., "Oligoaminonucleotiside Phosphoramidates, Oligomerization of Dimers of 3'-amino-3'-deoxy-nucleotides (GC and CG) in Aqueous Solution," Nuc. Acids Res. 15:1699:1715 (1987).
Zielinski, et al., "Oligomerization of Activated Derivatives of 3'-amino-3'-deoxyguanosine on poly(C) and poly(G) Templates," Nuc. Acids Res. 13:2469-2484 (1985).
Zielinski, et al., Autocatalytic Synthesis of a Tetranucleotide Analog, Nature 327:346 347 (1987). *
Zielinski, et al., Oligoaminonucleotiside Phosphoramidates, Oligomerization of Dimers of 3 amino 3 deoxy nucleotides (GC and CG) in Aqueous Solution, Nuc. Acids Res . 15:1699:1715 (1987). *
Zielinski, et al., Oligomerization of Activated Derivatives of 3 amino 3 deoxyguanosine on poly(C) and poly(G) Templates, Nuc. Acids Res . 13:2469 2484 (1985). *

Cited By (156)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6342214B1 (en) * 1995-05-16 2002-01-29 Karl Tryggvason Method for viral vector delivery
US6638264B1 (en) 1995-05-16 2003-10-28 Biostratum Incorporation Perfusion apparatus and methods for pharmaceutical delivery
US9096636B2 (en) 1996-06-06 2015-08-04 Isis Pharmaceuticals, Inc. Chimeric oligomeric compounds and their use in gene modulation
US7695902B2 (en) 1996-06-06 2010-04-13 Isis Pharmaceuticals, Inc. Oligoribonucleotides and ribonucleases for cleaving RNA
US7812149B2 (en) 1996-06-06 2010-10-12 Isis Pharmaceuticals, Inc. 2′-Fluoro substituted oligomeric compounds and compositions for use in gene modulations
US6503711B1 (en) * 1997-06-18 2003-01-07 Ulrich J. Krull Nucleic acid biosensor diagnostics
US20100173863A1 (en) * 1998-02-25 2010-07-08 Schinazi Raymond F 2-Fluoronucleosides
US9180138B2 (en) 1998-02-25 2015-11-10 University Of Georgia Research Foundation, Inc. 2′-fluoronucleosides
US6440723B1 (en) 1998-12-30 2002-08-27 Oligos Etc. Inc. Arrays with modified oligonucleotide and polynucleotide compositions
US7994145B2 (en) 1999-07-22 2011-08-09 Takeshi Imanishi Bicyclonucleoside analogues
US20070270370A1 (en) * 1999-07-22 2007-11-22 Sankyo Company, Limited Novel bicyclonucleoside analogues
US20030212032A1 (en) * 1999-09-10 2003-11-13 Geron Corporation Oligonucleotide N3'→P5' thiophosphoramidates: their synthesis and use
US6608036B1 (en) 1999-09-10 2003-08-19 Geron Corporation Oligonucleotide N3′→P5′ thiophosphoramidates: their synthesis and administration to treat neoplasms
US6835826B2 (en) 1999-09-10 2004-12-28 Geron Corporation Oligonucleotide N3′→P5′ Thiophosphoramidates: their synthesis and use
US20050049408A1 (en) * 1999-09-10 2005-03-03 Sergei Gryaznov Oligonucleotide N3'->P5' thiophosphoramidates: their synthesis and use
US7138383B2 (en) 1999-09-10 2006-11-21 Geron Corporation Treating cancer using an oligonucleotide N3′->N5′ thiophosphoramidate
US20070037770A1 (en) * 1999-09-10 2007-02-15 Sergei Gryaznov Oligonucleotide N3'-P5' thiophosphoramidates: their synthesis and use
US8906615B2 (en) 2000-01-21 2014-12-09 Geron Corporation 2′-arabino-fluorooligonucleotide N3′→P5′ phosphoramidates: their synthesis and use
US8377644B2 (en) 2000-01-21 2013-02-19 Geron Corporation 2′-arabino-fluorooligonucleotide N3′→P5′ phosphoramidates: their synthesis and use
US7321029B2 (en) * 2000-01-21 2008-01-22 Geron Corporation 2′-arabino-fluorooligonucleotide N3′→P5′ phosphoramidates: their synthesis and use
US20080171859A1 (en) * 2000-01-21 2008-07-17 Geron Corporation 2'-arabino-fluorooligonucleotide N3'-P5' phosphoramidates: their synthesis and use
US20040126752A1 (en) * 2000-01-21 2004-07-01 Sergei Gryaznov 2'-Arabino-fluorooligonucleotide n3'-->p5' phosphoramidates: their synthesis and use
US7989603B2 (en) 2000-01-21 2011-08-02 Geron Corporation 2′-arabino-fluorooligonucleotide N3′-P5′ phosphoramidates: their synthesis and use
US10358459B2 (en) 2000-01-21 2019-07-23 Geron Corporation 2′-arabino-fluorooligonucleotide N3′˜P5′ phosphoramidates: their synthesis and use
US9765109B2 (en) 2000-01-21 2017-09-19 Geron Corporation 2′-arabino-fluorooligonucleotide N3′→P5′ phosphoramidates: their synthesis and use
WO2001053307A1 (en) * 2000-01-21 2001-07-26 Geron Corporation 2'-arabino-fluorooligonucleotide n3'→p5'phosphoramidates: their synthesis and use
US20020137074A1 (en) * 2000-11-21 2002-09-26 Piunno Paul A.E. Selectivity of nucleic acid diagnostic and microarray technologies by control of interfacial nucleic acid film chemistry
US8604183B2 (en) 2002-11-05 2013-12-10 Isis Pharmaceuticals, Inc. Compositions comprising alternating 2′-modified nucleosides for use in gene modulation
US20050085435A1 (en) * 2003-01-02 2005-04-21 Niles Edward G. Novel inhibitors of poxvirus replication
WO2004060323A2 (en) * 2003-01-02 2004-07-22 The Research Foundation Of State University Of Newyork Novel inhibitors of poxvirus replication
WO2004060323A3 (en) * 2003-01-02 2008-01-03 Univ New York State Res Found Novel inhibitors of poxvirus replication
EP2314692A2 (en) 2003-06-02 2011-04-27 University of Massachusetts Methods and compositions for enhancing the efficacy and specificity of RNAi
EP3502252A1 (en) 2003-06-02 2019-06-26 University of Massachusetts Methods and compositions for controlling efficacy of rna silencing
US9404112B2 (en) 2003-09-09 2016-08-02 Geron Corporation Modified oligonucleotides for telomerase inhibition
US10196641B2 (en) 2003-09-09 2019-02-05 Geron Corporation Modified oligonucleotides for telomerase inhibition
US9657296B2 (en) 2003-09-09 2017-05-23 Geron Corporation Modified oligonucleotides for telomerase inhibition
US9388415B2 (en) 2003-09-09 2016-07-12 Geron Corporation Modified oligonucleotides for telomerase inhibition
US9388416B2 (en) 2003-09-09 2016-07-12 Geron Corporation Modified oligonucleotides for telomerase inhibition
EP2821085A2 (en) 2003-09-12 2015-01-07 University of Massachusetts RNA interference for the treatment of gain-of-function disorders
EP3760234A2 (en) 2003-09-12 2021-01-06 University of Massachusetts Rna interference for the treatment of gain-of-function disorders
US10655127B2 (en) 2003-11-04 2020-05-19 Geron Corporation RNA amidates and thioamidates for RNAi
US9133233B2 (en) 2003-11-04 2015-09-15 Geron Corporation RNA amidates and thioamidates for RNAi
US9822360B2 (en) 2003-11-04 2017-11-21 Geron Corporation RNA amidates and thioamidates for RNAi
WO2005054494A2 (en) 2003-11-26 2005-06-16 University Of Massachusetts Sequence-specific inhibition of small rna function
EP3427585A1 (en) 2003-11-26 2019-01-16 University of Massachusetts Sequence-specific inhibition of small rna function
US8569474B2 (en) 2004-03-09 2013-10-29 Isis Pharmaceuticals, Inc. Double stranded constructs comprising one or more short strands hybridized to a longer strand
WO2005089287A2 (en) 2004-03-15 2005-09-29 City Of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded rna
EP2514758A1 (en) 2004-03-15 2012-10-24 City of Hope Methods and compositions for the specific inhibition of gene expression by double-stranded RNA
US8394947B2 (en) 2004-06-03 2013-03-12 Isis Pharmaceuticals, Inc. Positionally modified siRNA constructs
EP3296312A1 (en) 2004-07-02 2018-03-21 Geron Corporation Synthesis of protected 3'-amino 5'-phosphoramidite nucleoside monomers
US10035815B2 (en) 2004-07-02 2018-07-31 Geron Corporation Synthesis of protected 3′-amino nucleoside monomers
US20090105468A1 (en) * 2004-07-02 2009-04-23 Gryaznov Sergei M Synthesis of Protected 3'-Amino Nucleoside Monomers
US8748593B2 (en) 2004-07-02 2014-06-10 Geron Corporation Synthesis of protected 3′-amino nucleoside monomers
US9365606B2 (en) 2004-07-02 2016-06-14 Geron Corporation Synthesis of protected 3′-amino nucleoside monomers
US10738073B2 (en) 2004-07-02 2020-08-11 Geron Corporation Synthesis of protected 3′-amino nucleoside monomers
US7884086B2 (en) 2004-09-08 2011-02-08 Isis Pharmaceuticals, Inc. Conjugates for use in hepatocyte free uptake assays
US20100029008A1 (en) * 2004-12-16 2010-02-04 Febit Biotech Gmbh Polymerase-independent analysis of the sequence of polynucleotides
US7968524B2 (en) 2007-05-15 2011-06-28 Helicon Therapeutics, Inc. Methods of enhancing long term memory formation by inhibition of Gpr12
US20090022667A1 (en) * 2007-05-15 2009-01-22 Marco Peters METHODS OF TREATING COGNITIVE DISORDERS BY INHIBITION OF Gpr12
US20100240441A1 (en) * 2007-09-14 2010-09-23 Konami Digital Entertainment Co., Ltd Game system, and game apparatus and challenge notifying apparatus constituting the game system
US10633654B2 (en) 2008-02-11 2020-04-28 Phio Pharmaceuticals Corp. Modified RNAi polynucleotides and uses thereof
US10131904B2 (en) 2008-02-11 2018-11-20 Rxi Pharmaceuticals Corporation Modified RNAi polynucleotides and uses thereof
US8815818B2 (en) 2008-07-18 2014-08-26 Rxi Pharmaceuticals Corporation Phagocytic cell delivery of RNAI
US10876119B2 (en) 2008-09-22 2020-12-29 Phio Pharmaceuticals Corp. Reduced size self-delivering RNAI compounds
US8796443B2 (en) 2008-09-22 2014-08-05 Rxi Pharmaceuticals Corporation Reduced size self-delivering RNAi compounds
US9175289B2 (en) 2008-09-22 2015-11-03 Rxi Pharmaceuticals Corporation Reduced size self-delivering RNAi compounds
US11396654B2 (en) 2008-09-22 2022-07-26 Phio Pharmaceuticals Corp. Neutral nanotransporters
US8664189B2 (en) 2008-09-22 2014-03-04 Rxi Pharmaceuticals Corporation RNA interference in skin indications
US10774330B2 (en) 2008-09-22 2020-09-15 Phio Pharmaceuticals Corp. Reduced size self-delivering RNAI compounds
US10041073B2 (en) 2008-09-22 2018-08-07 Rxi Pharmaceuticals Corporation Reduced size self-delivering RNAi compounds
US9303259B2 (en) 2008-09-22 2016-04-05 Rxi Pharmaceuticals Corporation RNA interference in skin indications
US9938530B2 (en) 2008-09-22 2018-04-10 Rxi Pharmaceuticals Corporation RNA interference in skin indications
US10138485B2 (en) 2008-09-22 2018-11-27 Rxi Pharmaceuticals Corporation Neutral nanotransporters
US10815485B2 (en) 2008-09-22 2020-10-27 Phio Pharmaceuticals Corp. RNA interference in skin indications
US9074211B2 (en) 2008-11-19 2015-07-07 Rxi Pharmaceuticals Corporation Inhibition of MAP4K4 through RNAI
US11254940B2 (en) 2008-11-19 2022-02-22 Phio Pharmaceuticals Corp. Inhibition of MAP4K4 through RNAi
US10167471B2 (en) 2009-01-05 2019-01-01 Rxi Pharmaceuticals Corporation Inhibition of PCSK9 through RNAI
US9493774B2 (en) 2009-01-05 2016-11-15 Rxi Pharmaceuticals Corporation Inhibition of PCSK9 through RNAi
US10479992B2 (en) 2009-02-04 2019-11-19 Phio Pharmaceuticals Corp. RNA duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
US9745574B2 (en) 2009-02-04 2017-08-29 Rxi Pharmaceuticals Corporation RNA duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
US11667915B2 (en) 2009-02-04 2023-06-06 Phio Pharmaceuticals Corp. RNA duplexes with single stranded phosphorothioate nucleotide regions for additional functionality
WO2010100404A2 (en) 2009-03-02 2010-09-10 Mina Therapeutics Limited Rna molecules and therapeutic uses thereof
WO2010105096A2 (en) 2009-03-11 2010-09-16 University Of Massachusetts Modulation of human cytomegalovirus replication by micro-rna 132 (mir132), micro-rna 145 (mir145) and micro-rna 212 (mir212)
WO2011075188A1 (en) 2009-12-18 2011-06-23 Dicerna Pharmaceuticals, Inc. Dicer substrate agents and methods for the specific inhibition of gene expression
US11584933B2 (en) 2010-03-24 2023-02-21 Phio Pharmaceuticals Corp. RNA interference in ocular indications
US9080171B2 (en) 2010-03-24 2015-07-14 RXi Parmaceuticals Corporation Reduced size self-delivering RNAi compounds
US11118178B2 (en) 2010-03-24 2021-09-14 Phio Pharmaceuticals Corp. Reduced size self-delivering RNAI compounds
US10184124B2 (en) 2010-03-24 2019-01-22 Phio Pharmaceuticals Corp. RNA interference in ocular indications
US9963702B2 (en) 2010-03-24 2018-05-08 Rxi Pharmaceuticals Corporation RNA interference in dermal and fibrotic indications
US10662430B2 (en) 2010-03-24 2020-05-26 Phio Pharmaceuticals Corp. RNA interference in ocular indications
US10240149B2 (en) 2010-03-24 2019-03-26 Phio Pharmaceuticals Corp. Reduced size self-delivering RNAi compounds
US9340786B2 (en) 2010-03-24 2016-05-17 Rxi Pharmaceuticals Corporation RNA interference in dermal and fibrotic indications
US10913948B2 (en) 2010-03-24 2021-02-09 Phio Pharmaceuticals Corp. RNA interference in dermal and fibrotic indications
WO2012129363A2 (en) 2011-03-24 2012-09-27 President And Fellows Of Harvard College Single cell nucleic acid detection and analysis
WO2013075132A1 (en) 2011-11-17 2013-05-23 The United States Of America, As Represented By The Secretary, Department Of Health & Human Services Therapeutic rna switches compositions and methods of use
US11401543B2 (en) * 2012-09-27 2022-08-02 Bioo Scientific Corporation Methods and compositions for improving removal of ribosomal RNA from biological samples
US9771581B2 (en) 2012-10-26 2017-09-26 Geron Corporation C-myc antisense oligonucleotides and methods for using the same to treat cell-proliferative disorders
US9228189B2 (en) 2012-10-26 2016-01-05 Geron Corporation C-myc antisense oligonucleotides and methods for using the same to treat cell-proliferative disorders
WO2014113540A1 (en) 2013-01-16 2014-07-24 Iowa State University Research Foundation, Inc. A deep intronic target for splicing correction on spinal muscular atrophy gene
EP4012031A1 (en) 2013-07-03 2022-06-15 Dicerna Pharmaceuticals, Inc. Methods and compositions for the specific inhibition of alpha-1 antitrypsin by double-stranded rna
EP3444350A1 (en) 2013-07-03 2019-02-20 Dicerna Pharmaceuticals, Inc. Methods and compositions for the specific inhibition of alpha-1 antitrypsin by double-stranded rna
US10934550B2 (en) 2013-12-02 2021-03-02 Phio Pharmaceuticals Corp. Immunotherapy of cancer
US11279934B2 (en) 2014-04-28 2022-03-22 Phio Pharmaceuticals Corp. Methods for treating cancer using nucleic acids targeting MDM2 or MYCN
US9796747B2 (en) 2014-05-01 2017-10-24 Geron Corporation Oligonucleotide compositions and methods of making the same
US11739114B2 (en) 2014-05-01 2023-08-29 Geron Corporation Oligonucleotide compositions and methods of making the same
US11299511B2 (en) 2014-05-01 2022-04-12 Geron Corporation Oligonucleotide compositions and methods of making the same
US10392418B2 (en) 2014-05-01 2019-08-27 Geron Corporation Oligonucleotide compositions and methods of making the same
US10900039B2 (en) 2014-09-05 2021-01-26 Phio Pharmaceuticals Corp. Methods for treating aging and skin disorders using nucleic acids targeting Tyr or MMP1
US11926828B2 (en) 2014-09-05 2024-03-12 Phio Pharmaceuticals Corp. Methods for treating aging and skin disorders using nucleic acids targeting TYR or MMP1
US10556020B2 (en) 2014-09-26 2020-02-11 University Of Massachusetts RNA-modulating agents
US11464873B2 (en) 2014-09-26 2022-10-11 University Of Massachusetts RNA-modulating agents
EP3865576A1 (en) 2014-12-15 2021-08-18 Dicerna Pharmaceuticals, Inc. Ligand-modified double-stranded nucleic acids
EP3569711A1 (en) 2014-12-15 2019-11-20 Dicerna Pharmaceuticals, Inc. Ligand-modified double-stranded nucleic acids
WO2016100401A1 (en) 2014-12-15 2016-06-23 Dicerna Pharmaceuticals, Inc. Ligand-modified double-stranded nucleic acids
WO2016161378A1 (en) 2015-04-03 2016-10-06 University Of Massachusetts Oligonucleotide compounds for treatment of preeclampsia and other angiogenic disorders
WO2016161388A1 (en) 2015-04-03 2016-10-06 University Of Massachusetts Fully stabilized asymmetric sirna
US10435688B2 (en) 2015-04-03 2019-10-08 University Of Massachusetts Oligonucleotide compounds for targeting huntingtin mRNA
EP3766973A1 (en) 2015-04-03 2021-01-20 University Of Massachusetts Oligonucleotide compounds for targeting huntingtin mrna
US11345917B2 (en) 2015-04-03 2022-05-31 University Of Massachusetts Oligonucleotide compounds for treatment of preeclampsia and other angiogenic disorders
US10774327B2 (en) 2015-04-03 2020-09-15 University Of Massachusetts Oligonucleotide compounds for targeting huntingtin mRNA
US9862952B2 (en) 2015-04-03 2018-01-09 University Of Massachusetts Oligonucleotide compounds for treatment of preeclampsia and other angiogenic disorders
EP3995581A2 (en) 2015-04-03 2022-05-11 University of Massachusetts Oligonucleotide compounds for treatment of preeclampsia and other angiogenic disorders
WO2016161374A1 (en) 2015-04-03 2016-10-06 University Of Massachusetts Oligonucleotide compounds for targeting huntingtin mrna
US10519451B2 (en) 2015-04-03 2019-12-31 University Of Massachusetts Oligonucleotide compounds for treatment of preeclampsia and other angiogenic disorders
US9809817B2 (en) 2015-04-03 2017-11-07 University Of Massachusetts Oligonucleotide compounds for targeting huntingtin mRNA
US11230713B2 (en) 2015-04-03 2022-01-25 University Of Massachusetts Oligonucleotide compounds for targeting huntingtin mRNA
EP3929293A2 (en) 2015-04-03 2021-12-29 University Of Massachusetts Fully stabilized asymmetric sirna
EP3708680A1 (en) 2015-04-15 2020-09-16 The University of Massachusetts Compositions and methods for xi chromosome reactivation
US10745687B2 (en) 2015-04-23 2020-08-18 Geron Corporation Methods of polynucleotide preparation using multivalent cation salt compositions
US11441144B2 (en) 2015-04-23 2022-09-13 Geron Corporation Methods of polynucleotide preparation using multivalent cation salt compositions
WO2016172346A1 (en) 2015-04-23 2016-10-27 Geron Corporation Methods of polynucleotide preparation using multivalent cation salt compositions
US10689647B2 (en) 2015-05-08 2020-06-23 Dicerna Pharmaceuticals, Inc. Methods and compositions for the specific inhibition of antithrombin 3 (AT3) by double-stranded RNA
US11667918B2 (en) 2015-05-08 2023-06-06 Dicerna Pharmaceuticals, Inc. Methods and compositions for the specific inhibition of antithrombin 3 (AT3) by double-stranded RNA
US10808247B2 (en) 2015-07-06 2020-10-20 Phio Pharmaceuticals Corp. Methods for treating neurological disorders using a synergistic small molecule and nucleic acids therapeutic approach
US11001845B2 (en) 2015-07-06 2021-05-11 Phio Pharmaceuticals Corp. Nucleic acid molecules targeting superoxide dismutase 1 (SOD1)
WO2017015075A1 (en) 2015-07-17 2017-01-26 President And Fellows Of Harvard College Methods of amplifying nucleic acid sequences
US10633653B2 (en) 2015-08-14 2020-04-28 University Of Massachusetts Bioactive conjugates for oligonucleotide delivery
WO2017049192A1 (en) 2015-09-17 2017-03-23 University Of Massachusetts Compositions and methods for modulating fmr1 expression
US11021707B2 (en) 2015-10-19 2021-06-01 Phio Pharmaceuticals Corp. Reduced size self-delivering nucleic acid compounds targeting long non-coding RNA
US10799591B2 (en) 2016-01-31 2020-10-13 University Of Massachusetts Branched oligonucleotides
US11896669B2 (en) 2016-01-31 2024-02-13 University Of Massachusetts Branched oligonucleotides
US10478503B2 (en) 2016-01-31 2019-11-19 University Of Massachusetts Branched oligonucleotides
US11753638B2 (en) 2016-08-12 2023-09-12 University Of Massachusetts Conjugated oligonucleotides
EP3896451A1 (en) 2016-11-07 2021-10-20 University Of Massachusetts Carm1 inhibitors for facioscapulohumeral muscular dystrophy
US10443055B2 (en) 2016-12-22 2019-10-15 Beth Israel Deaconess Medical Center Compounds that target MYC microRNA responsive elements for the treatment of MYC-associated cancer
US11401519B2 (en) 2017-06-07 2022-08-02 University Of Massachusetts Anti-ADAM33 oligonucleotides and related methods
US11306312B2 (en) 2017-09-01 2022-04-19 Thomas Jefferson University Compositions and methods for MYC messenger RNA inhibitors
US11827882B2 (en) 2018-08-10 2023-11-28 University Of Massachusetts Modified oligonucleotides targeting SNPs
US11629347B2 (en) 2019-05-06 2023-04-18 University Of Massachusetts Anti-C9ORF72 oligonucleotides and related methods
US11873487B2 (en) 2019-06-20 2024-01-16 University Of Massachusetts Compositions and methods for improved gene editing
US11702659B2 (en) 2021-06-23 2023-07-18 University Of Massachusetts Optimized anti-FLT1 oligonucleotide compounds for treatment of preeclampsia and other angiogenic disorders
WO2023086295A2 (en) 2021-11-10 2023-05-19 University Of Rochester Antisense oligonucleotides for modifying protein expression
WO2023086292A2 (en) 2021-11-10 2023-05-19 University Of Rochester Gata4-targeted therapeutics for treatment of cardiac hypertrophy
WO2023164636A1 (en) 2022-02-25 2023-08-31 Vor Biopharma Inc. Compositions and methods for homology-directed repair gene modification
WO2023196816A1 (en) 2022-04-04 2023-10-12 Vor Biopharma Inc. Compositions and methods for mediating epitope engineering
WO2024050261A1 (en) 2022-08-29 2024-03-07 University Of Rochester Antisense oligonucleotide-based anti-fibrotic therapeutics

Also Published As

Publication number Publication date
ATE212997T1 (en) 2002-02-15
DE69619141D1 (en) 2002-03-21
PL183661B1 (en) 2002-06-28
PL328639A1 (en) 1999-02-15
WO1997031009A1 (en) 1997-08-28
DE69619141T2 (en) 2002-09-26
AU6178996A (en) 1997-09-10
EP0882059B1 (en) 2002-02-06
AU703509B2 (en) 1999-03-25
CZ262998A3 (en) 1999-05-12
EP0882059A1 (en) 1998-12-09
US5824793A (en) 1998-10-20

Similar Documents

Publication Publication Date Title
US5684143A (en) Oligo-2'-fluoronucleotide N3'->P5' phosphoramidates
EP0549615B1 (en) Sugar modified oligonucleotides that detect and modulate gene expression
US5473060A (en) Oligonucleotide clamps having diagnostic applications
US5614617A (en) Nuclease resistant, pyrimidine modified oligonucleotides that detect and modulate gene expression
US7572582B2 (en) Oligonucleotide analogues
US5670633A (en) Sugar modified oligonucleotides that detect and modulate gene expression
EP1015469B2 (en) Bi- and tri-cyclic nucleoside, nucleotide and oligonucleoide analogues
US6794499B2 (en) Oligonucleotide analogues
US5571903A (en) Auto-ligating oligonucleotide compounds
JPH03190895A (en) Nucleotide modification compound
US5859233A (en) Synthons for synthesis of oligonucleotide N3-P5 phosphoramidates
JPH10503773A (en) 5'-dithio modified oligonucleotide
WO1997037691A1 (en) Telomerase inhibitors
EP0931090B1 (en) Improved chimeric oligonucleotide vectors
JP4180681B2 (en) Antisense oligonucleotide
EP1228243A1 (en) Compositions and methods of synthesis and use of novel nucleic acid structures

Legal Events

Date Code Title Description
STCF Information on status: patent grant

Free format text: PATENTED CASE

AS Assignment

Owner name: LYNX THERAPEUTICS, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GRYAZNOV, SERGEI M.;SCHULTZ, RONALD G.;REEL/FRAME:009038/0260;SIGNING DATES FROM 19970627 TO 19971126

FEPP Fee payment procedure

Free format text: PAYOR NUMBER ASSIGNED (ORIGINAL EVENT CODE: ASPN); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

FPAY Fee payment

Year of fee payment: 4

AS Assignment

Owner name: GERON CORPORATION, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:LYNX THERAPEUTICS, INC.;REEL/FRAME:012831/0709

Effective date: 20020402

FPAY Fee payment

Year of fee payment: 8

FPAY Fee payment

Year of fee payment: 12